Archive for the ‘Bone Marrow Stem Cells’ Category
Cancer Patients Who Need Stem Cell Transplants May Have New Donor Options – Everyday Health
For some people with blood cancers who need a stem cell transplant, finding a donor who is an excellent match can mean the difference between life and death.
Unfortunately, even though there are more than 40 million potential donors in the national registry, finding a perfect match isnt always possible, especially in underrepresented racial and ethnic groups.
But a new approach using an old chemotherapy drug,cyclophosphamide, isis opening up new possibilities for people with cancers like leukemia, lymphoma, and multiple myeloma. Researchers have found that by administering the drug several days after transplantation, people receiving blood stem cells from unrelated, partially matched donors can have survival rates comparable with those who received exactly matched cells.
[1]
This innovative approach can greatly expand patient access to safe and effective stem cell transplant, regardless of matching degree with the donor, says lead coauthor Monzr M. Al Malki, MD, a hematologist and oncologist and director of the Unrelated Donor BMT program at City of Hope, a cancer research and treatment organization with locations across the United States.
Thats exciting because it means more patients will be able to receive this potentially life-extending therapy, says Dr. Al Malki.
Donor compatibility is determined by a set of protein markers on blood cells called HLAs (human leukocyte antigens), says David Miklos, MD, a professor of medicine and chief of Stanford BMT and Cell Therapy Program at Stanford Medicine in California. Stanford was one of the medical sites of the trial, though Dr. Miklos is not a coauthor of the research.
[2]
[3]
Why was an exact match needed? Anything less increased the likelihood of a graft failure, as well as graft-versus-host disease meaning the transplanted cells attack the patients own, which can cause serious or even fatal complications, explains Miklos.
About a decade ago, researchers started using cyclophosphamide to destroy the parts of a persons immune system that would reject the transplant. That breakthrough allowed researchers to not only have better outcomes in fully matched donors, it also opened the door for successful transplants between people who were only partial matches.
[4]
The new study looked at cyclophosphamide treatment in patients receiving peripheral blood stem cell transplantation meaning healthy stem cells are harvested from a donors bloodstream, and then administered via infusion to the person with cancer.
Blood stem cell transplantation has largely replaced bone marrow transplantation, according to researchers.It's an easier way of collecting stem cells from donors, and its a little safer, because donors dont need to be under anesthesia as they would in bone marrow transplantation, says Al Malki.
For this part of the study, the researchers examined data from 70 adults who were 65 years old on average, all with advanced blood cancers. Participants received a reduced-intensity conditioning regimen to somewhat suppress their immune system to prepare them for transplantation, followed by an infusion stem cells from unrelated, partially matched donors.
The researchers reported an overall high survival rate of 79 percent at one year which is comparable to survival rates seen with fully matched donors.
The main side effect or risk of transplantation is graft-versus-host disease, says Al Malki. After one year, 51 percent of participants were free of the disease and had not relapsed, which is also comparable to what would be seen with fully matched donors, he says.
Historically, barriers in access to transplant have existed due to the low availability of matched, related sibling donors, as well as the substantial variance of matched, unrelated donor availability, especially for patients with diverse ancestry, says study coauthor Steven M. Devine, MD, chief medical officer of NMDP (formerly known as the National Marrow Donor Program and Be The Match).
These findings advance our ability to offer more options to patients without a fully matched donor, many of whom are ethnically diverse and have been underserved in receiving potentially lifesaving cell therapy, says Dr. Devine.
These findings are incredibly important and critical in the effort to improve existing inequities, says Miklos.
In the past, we could not bring some patients forward to receive this lifesaving therapy because they didnt have a compatible donor, but with the new approach of using post-transplant cyclophosphamide, all patients have donors now, he says.
See the rest here:
Cancer Patients Who Need Stem Cell Transplants May Have New Donor Options - Everyday Health
Efficacy and safety of stem cell transplantation for multiple sclerosis: a systematic review and meta-analysis of … – Nature.com
Study selection
From the initial literature search, we retrieved relevant 3948 records from PubMed, Web of Science, Scopus, and the Cochrane Library. After the title and abstract screening of them we screened the full text of 295 articles. Only nine studies met our criteria31,32,33,34,35,36,37,38,39. Figure1 shows the PRISMA flow diagram of our search and selection process.
The nine studies were RCTs and enrolled a total of 422 multiple sclerosis patients. All studies were parallel in design except 4 studies were cross-over RCTs31,32,34,39. These cross-over trials were reviewed up to the point of cross-over. All studies infused stem cells intravenously except Petrou et al. that included an additional intrathecal SCT subgroup32. This study showed that intrathecal SCT was more effective than intravenous SCT, but we pooled the data of both routes as single study data. Of the included studies, only two studies used autologous hematopoietic SCT (AHSCT) in addition to immune ablative regimen prior to the transplantation37,38. Burt et al. compared SCT to DMTs (natalizumab, fingolimod, and dimethyl fumarate) in RRMS patients37, and Mancardi et al., compared SCT to mitoxantrone in relapsing and progressive MS patients38.
Supplementary Table S1 summarizes the characteristics of the included trials, Table 1 shows the demographic and baseline characteristics of these studies population, and Supplementary Table S3 shows efficacy endpoints reported at 6months.
We assessed seven domains in each study according to The Cochrane Collaborations tool for assessing risk of bias 1. The 9 studies were randomized but 4 studies32,35,38,39 didnt clarify the methods of random sequence generation. 6 RCTs confirmed concealment of patients allocation to the intervention31,32,33,34,36,37. Blinding of the outcome assessors was clearly stated in all studies except Nabavi et al.39 but blinding of participants and personnel wasnt fulfilled in three studies35,37,38. The reasons for incomplete outcome data are related to the treatment in Uccelli et al.31 and the reasons werent clearly described in Burt et al37. Two studies reported the outcomes in an incomplete way that limited their inclusion in the meta-analysis inducing a reporting bias33,38. The overall quality of the studies was good for 2 studies32,36, fair for 3 studies31,34,37, and poor for 4 studies33,35,38,39. Figure2 shows the risk of bias summary and graph.
Risk of bias assessment: (a) Risk of bias summary, (b) Risk of bias graph.
After analyzing the efficacy and safety outcomes for all studies collectively, we subdivided the results into studies that used immunosuppression before AHSCT37,38 and studies that transplanted mesenchymal stem cells (MSCs) without immunosuppression31,32,33,34,35,36,39 to minimize the procedural variations among the included trials.
The majority of the studies31,32,33,34,35,36,37,39 reported EDSS change for 211 patients in stem cell transplantation (SCT) arm and 176 controls. Because the time of reporting this outcome varied among the studies, we analyzed EDSS change at the last follow-up reported by each study. Our analysis showed nonsignificant difference between SCT group and the control group (MD=0.48, 95% CI [1.11, 0.14], p=0.13). There was great heterogeneity between studies (2=116.74, df=7, p<0.00001, I2=94%), so we pooled the data under the random-effects model (Table 2 and Supplementary Figure S1).
The subgroup analysis of the studies that used MSCs without immunosuppression also showed nonsignificant improvement (MD=0.3, 95% CI [0.87, 0.27], p=0.3). However, Burt et al37. that used immunosuppression before AHSCT revealed significant EDSS reduction (Supplementary Figure S1).
The results remained nonsignificant after the leave-one-out sensitivity analysis (Supplementary Figure L1).
The heterogeneity within the studies was not significant (2=1.61, df=1, p=0.2, I2=38%), and we adopted a random effect model. The reduction of EDSS in SCT group was significantly greater than the control group (MD=0.57, 95% CI [1.08, 0.06], p=0.03) (Table 2 and Fig.3a).
Forest plot of EDSS change from baseline at (a) 2months, (b) 6months, (c) 12months.
Adopting the random-effects model, the heterogeneity between the studies was significant (2=65.27, df=6, p<0.00001, I2=91%), and SCT showed nonsignificant improvement of EDSS compared to the control (MD=0.48, 95% CI [0.98, 0.03], p=0.07) (Table 2 and Fig.3b). MSCs without immunosuppression also resulted in nonsignificant EDSS reduction at 6months (MD=0.33, 95% CI [0.78, 0.11], p=0.14) (Fig.3b).
The effect estimate changed to (MD=0.62,95% CI [1.14, 0.09], p=0.02) favoring SCT over the control after excluding Nabavi et al.39 from the analysis (Supplementary Figure L2 and Table L1).
We adopted the random-effects model because heterogeneity was significant, and the difference between the SCT group and the control was not significant at 12months for both collective studies analysis and studies used MSCs without immunosuppression (p=0.06 and p=0.5, respectively). However, the study that used AHSCT plus immunosuppression37 showed significant improvement in patients disability (p<0.00001) (Table 2 and Fig.3c).
After performing a sensitivity analysis by excluding Fernandez et al.36, the results changed from nonsignificant to significant improvement in SCT arm (MD=1.69, 95% CI [1.94, 1.44], p<0.00001) (Supplementary Figure L3 and Table L1).
We compared the effect of SCT on patients disability depending on baseline EDSS. Six studies31,32,33,34,37,39 included 334 MS patients with baseline EDSS6.5, while two studies35,36 included 53 patients with baseline EDSS>6.5. Using a random effects model, both subgroups showed significant heterogeneity (p<0.00001 and p<0.00001). Both subgroups revealed nonsignificant effect of SCT on EDSS, (MD=0.41, 95% CI [1.11, 0.29], p=0.25) for baseline EDSS6.5 subgroup and (MD=0.68, 95%CI [2.68, 1.32], p=0.5) for baseline EDSS>6.5 subgroup (Table 2 and Supplementary Figure S2).
We pooled data of EDSS change from baseline to the last assessment time under a random-effects model, and the differences were nonsignificant for both low and high doses subgroups, (MD=0.31, 95% CI [1, 0.38], p=0.37) and (MD=0.57, 95% CI [1.94, 0.8], p=0.41), respectively. The studies of both subgroups showed significant heterogeneity (I2=95%, p<0.00001) for the low doses subgroup, and (I2=89%, p=0.0001) for the high doses subgroup (Table 2 and Supplementary Figure S3).
Adopting a random-effects model, stem cells from embryonic as well as adult origin showed nonsignificant effect on EDSS (p=0.17, and p=0.37, respectively), With significant heterogeneity among the studies (I2=88%, p=0.004), and (I2=94%, p<0.00001), respectively (Table 2 and Supplementary Figure S4).
We pooled data using a random-effects model. Five studies31,32,33,36,39, in which placebo was the control, showed substantialheterogeneity (I2=63%, p=0.03) and the difference between SCT and placebo was not significant (MD=0.09, 95% CI [0.46, 0.28], p=0.62). Three studies34,35,37, in which the control was active treatment, showed significant reduction of EDSS with SCT compared to the active drugs (MD=1.21, 95% CI [1.98, 0.43], p=0.002) and the heterogeneity was significant (I2=88%, p=0.0002) (Table 2 and Supplementary Figure S5).
Only two studies32,34 reported the number of relapses in the 6months following the intervention. Under a random-effects model, the heterogeneity was moderate (p=0.14, I2=53%), and the decrease in relapses number was nonsignificant (p=0.23) (Supplementary Figure S6).
Four studies31,32,34,37 assessed T25-FW in 154 and 136 patients in the SCT and control groups, respectively. We pooled data under a random-effect model, and heterogeneity was moderate (2=5.99, df=3, p=0.11, I2=50%). SCT resulted in a nonsignificant improvement in patients T25-FW scores compared to the control group (MD=0.69, 95% CI [1.93, 0.56], p=0.28), as shown in Fig.4.
Forest plot of T25-FW change from baseline.
In the studies that included mesenchymal SCT without immunosuppression, the improvement in patients T25-FW scores after SCT was not significant (MD=0.39, 95% CI [0.84, 0.06], p=0.09), but T25-FW significantly improved in the study that used AHSCT and immunosuppression37 (p=0.006). Figure4 demonstrates these analyses. The p value of the results didnt change after the one-study-removed sensitivity analysis (Supplementary Figure L4).
9-HPT was evaluated in four RCTs31,32,34,37. We used a random-effects model because heterogeneity was significant (p=0.0003, I2=84%). 9-HPT showed nonsignificant improvement in the collective analysis and the sub-analysis of MSCs without immunosuppression. However, Burt et al.37. revealed a significant improvement (p<0.00001) (Supplementary Figure S7). The results remained nonsignificant after sensitivity analysis (Supplementary Figure L5).
We pooled PASAT-3 scores assessed at the end of treatment in four trials under a random-effects model31,34,36,37. Heterogeneity was minimal (p=0.35, I2=9%), and the differences were nonsignificant in the collective analysis and the sub-analysis of autologous and mesenchymal SCT (p=0.35, p=0.96, and p=0.31, respectively) (Supplementary Figure S8). Effect estimate remained nonsignificant after one-study-removed sensitivity analysis (Supplementary Figure L6).
We analyzed the change in brain lesion volume from baseline to the end of the follow-up. Data were pooled under a random-effects model, heterogeneity was absent (p=0.38, I2=0%). Our analysis revealed a significant reduction in T2 lesions volume (MD=7.05, 95% CI [10.69, 3.4], p=0.0002). In the studies that used MSCs without immunosuppression, the reduction of brain lesions volume was nonsignificant (p=0.1) (Fig.5a).
Forest plot of radiological outcomes change from baseline (a) MRI T2-weighted lesions volume at the end of treatment, (b) MRI T2-weighted lesions number at 12months, (c) number of GELs at the end of treatment. *the study used immunosuppression before AHSCT.
The results became nonsignificant and changed to (MD=4.41, 95% CI [9.66, 0.85], p=0.1) after a sensitivity analysis performed by excluding Burt et al.37 (Supplementary Figure L7 and Table L1).
Adopting a random-effects model, the studies showed substantial heterogeneity (p=0.07, I2=70%). And the differences between SCT and the control after 12months were nonsignificant (p=0.99) (Fig.5b).
Five studies31,32,33,34,36 assessed this outcome. Four studies reported the change of GELs number from baseline at 6months except Fernandez et al.36 at 12months. We pooled data under a random-effects model and heterogeneity was not significant (2=7.81, df=4, p=0.1, I2=49%). Our analysis revealed nonsignificant differences in GELs number change (p=0.83) (Fig.5c). The results didnt change after sensitivity analysis (Supplementary Figure L8).
Seven studies31,32,33,34,36,37,38 reported adverse events that occurred during the follow-up period. Two studies35,39 didnt provide data about AEs. Nabavi et al. mentioned only pain at the site of bone marrow aspiration39. Our analysis revealed that the difference was nonsignificant between SCT and the control group regarding the incidence of most AEs. Administration-related AEs, including infusion site swelling, hematoma, and pain, were significantly more common in the SCT group compared to the control (N=25, RR=2.55, 95% CI [1.08, 6.03], p=0.034). On the other hand, the SCT group had a lower incidence of total infections (any infection during the follow-up period, including viral infections, respiratory, urinary infections, scabies, and other infestations) than the control group (N=60, RR=0.58, 95% CI [0.37, 0.9], p=0.02). Regarding the use of immunosuppression, AHSCT combined with immunosuppression was significantly associated with a higher incidence of blood and lymphatic system disorders (N=16, RR=2.33, 95% CI [1.23, 4.39], p=0.009). The analyses of the adverse events are shown in Table 3 and Supplementary Figures S9S14. No transplant-related mortality was noted in all trials during the follow-up period, except for two unrelated deaths compacted by Fernandez et al. in the placebo arm (one due to choking while feeding and the other due to respiratory infection)36.
We examined the publication bias among the studies that reported the effect of SCT on patients disability using the funnel plot test. Although there was funnel plot asymmetry, the test isnt reliable because the included studies were less than ten studies24 (Supplementary Figure S15).
Read the original here:
Efficacy and safety of stem cell transplantation for multiple sclerosis: a systematic review and meta-analysis of ... - Nature.com
All-in-one Hangzhou Protocol: killing four birds with one stone | Cell Research – Nature.com
In a recent study published in The New England Journal of Medicine, researchers developed an innovative all-in-one treatment protocol that combines CD7 chimeric antigen receptor T-cell therapy with subsequent haploidentical hematopoietic stem cell transplantation. This approach significantly advances the treatment of CD7+ hematological malignancies by eliminating the need for myeloablative chemotherapy and immunosuppressants for graft-versus-host disease prophylaxis.
Chimeric antigen receptor (CAR) T-cell therapy utilizes synthetic antigen receptors targeting T lymphocytes to attack tumor cells specifically. To date, CAR T-cell therapies have shown unprecedented efficacy in B-cell malignancies. In contrast, there are significantly fewer clinical trials of CAR T-cell therapy against T-cell malignancies.1 Outcomes of T-cell lymphomas and relapsed T-cell acute lymphoblastic leukemia (T-ALL) patients are notably poor compared to those of their B-cell counterparts, with an estimated 5-year overall survival rate of only 32% for T-cell lymphomas patients and 7% for relapsed T-ALL patients.2 The challenge in treating T-cell malignancies lies in the lack of ideal target antigens. Several studies, including a recent study published in Cell Research,3 identified CD7 as a promising target for CAR-T therapy in the treatment of T-cell malignancies. However, in these clinical trials of CD7 CAR T-cells, researchers frequently observed that CD7 CAR T-cell treatment resulted in incomplete hematologic recovery and pancytopenia, presenting a significant clinical challenge.4
Recently, The New England Journal of Medicine reported on a novel all-in-one treatment regimen developed by a team from Hangzhou, China (hereafter referred to as the Hangzhou Protocol).5 In this clinical trial of CD7 CAR T-cell therapy for CD7+ hematological malignancies, including T-cell malignancies and acute myeloid leukemia (AML), Hu et al. observed that CD7 CAR T-cell treatment induced severe bone marrow hypocellularity, pancytopenia, and immunosuppression, aligning with previous reports. The first patient experienced persistent grade 4 pancytopenia for three months following CAR T-cell infusion and consequently developed severe infections. A salvage haploidentical hematopoietic stem cell transplantation (HSCT) was thus performed, without additional pharmacologic pre-HSCT conditioning regimens. Favorable engraftment of hematopoietic stem cells (HSCs) and immune reconstitution were observed, suggesting that the potent immunosuppressive functions of CD7 CAR T-cells, coupled with prior lymphodepletion, might replace the need for traditional chemotherapy conditioning. The subsequent 9 patients of similar conditions were promptly subjected to HSCT within one month after receiving CD7 CAR T-cell therapy without chemotherapy conditioning and immunosuppressive drugs for graft-versus-host disease (GVHD) prophylaxis. All 10 patients treated with the Hangzhou Protocol achieved complete remission. Pancytopenia was successfully relieved after allogeneic HSCT. Eight patients had full donor chimerism and immune reconstitution with a mild and manageable incidence of acute GVHD. Six of these patients remained in minimal residual disease-negative complete remission. The one-year overall survival rate increased to 68%. Collectively, this all-in-one Hangzhou Protocol offers a promising therapeutic option to CD7+ tumor patients.
Traditional CAR T-cell therapy followed by bridging HSCT involves longer intervals, chemotherapy conditioning, and the use of immunosuppressants for GVHD prophylaxis. Notably, the Hangzhou Protocol offers quadruple clinical benefits (Fig.1). First, CD7 CAR T-cell therapy efficiently eliminates CD7+ cancer cells in patients. More than 95% of patients with T-cell malignancies have CD7+ tumor cells,6 and about 30% of AML patients exhibit CD7 expression.7 In the trial, all treated patients including those with both T-cell malignancies and AML achieved complete remission. Second, the Protocol eliminates the need for pharmacologic myeloablation before HSCT, thereby avoiding its associated toxic effects. This is particularly beneficial for patients who have severe physiological issues or are in poor condition and who often are ineligible for allogeneic HSCT. Third, the Hangzhou Protocol does not require the use of immunosuppressants for GVHD prophylaxis, thus avoiding subsequent immunodeficiency issues. This allows the patients immune system to recover more rapidly. However, the mechanism behind this benefit requires further investigation. It is reported that CD7 plays a costimulatory role in T-cell signaling.8 Therefore, CD7 T cells, which are generated post CD7 CAR T-cell treatment, display reduced functionality and consequently a lower degree of GVHD. Lastly, the all-in-one treatment regimen maintains the persistence of CAR T-cells and supports their long-term immune surveillance function along with the graft-versus-leukemia effect. This maximizes the benefits of long-term immune surveillance by CAR T-cells and minimizes the risk of tumor relapse.
The figure illustrates clinical benefits of the Hangzhou Protocol in the context of CD7 CAR T-cell therapy for hematological malignancies. The top left section highlights the targeted tumor clearance capability of CD7 CAR T-cells, which are effective against both T-cell malignancies and AML. The top right section demonstrates the elimination of the need for myeloablative chemotherapy, reducing patient exposure to toxic effects. The bottom left section shows the avoidance of immunosuppressants in GVHD prophylaxis, which not only facilitates quicker immune system recovery but also minimizes complications from immunosuppression and associated drug side effects. Finally, the bottom right section details the sustained CAR T and GVL effects, which are crucial for preventing tumor relapse and supporting long-term immune surveillance. GVL graft-versus-leukemia, BM bone marrow, GVHD graft-versus-host disease, AML acute myeloid leukemia, CAR chimeric antigen receptor.
Several questions remain to be addressed regarding CD7 CAR T-cell therapy. One interesting question is why CD7 CAR T-cells suppress the patients hematopoiesis, causing severe pancytopenia, bone marrow aplasia, and immunosuppression. CAR T-cell-induced cytokine release syndrome could induce cytokine-associated pancytopenia.9 Conversely, it is also possible that minimal expression of CD7 on HSCs might induce CAR T-cell cytotoxicity. Furthermore, the mechanisms behind the successful engraftment and immune reconstitution of donor-derived stem cells in the presence of CD7 CAR T-cells while the patients own hematopoiesis is suppressed, are intriguing and warrant further investigation. Another interesting point is why CD7 CAR T-cell persistence is significantly longer than that of CD19 CAR T-cells. CAR tonic signaling has been reported to play a crucial role in regulating in vivo persistence and fitness of CAR T-cells.10,11 Thus, it is necessary to explore whether CD7 knockout in CD7 CAR T-cells affects CAR tonic signaling. All these questions need to be addressed in future studies to enhance our understanding and improve the efficacy of CD7 CAR T-cell therapy.
More here:
All-in-one Hangzhou Protocol: killing four birds with one stone | Cell Research - Nature.com
Abu Dhabi Stem Cells Center first healthcare institution in UAE to receive FACT accreditation for cellular therapy … – Abu Dhabi Media Office
Abu Dhabi Stem Cells Center (ADSCC) has received the internationally recognised The Foundation for the Accreditation of Cellular Therapy (FACT) accreditation for its comprehensive cellular therapy programme.
Under the leadership of its Chief Executive Officer Prof. Yendry Ventura, ADSCC is the first healthcare institution in the UAE to be recognised by FACT, positioning the UAE as a leading destination for cutting-edge medical advancements in the region.
Out of the 261 centres worldwide accredited by FACT for cellular therapy, ADSCC stands as one of only two centres in the entire Middle East to meet FACTs rigorous standards. This accomplishment solidifies ADSCC's position as a leader in advanced cellular therapy. The centre achieved FACT accreditation for FACT-JACIE International Standards for Hematopoietic Cellular Therapy Product Processing, which implement best quality standards of medical and laboratory practice in hematopoietic progenitor cell transplantation and therapies from any hematopoietic tissue source (marrow, peripheral blood, umbilical cord and placental blood).
In 2020, ADSCC launched its Abu Dhabi Bone Marrow Transplant programme, the first comprehensive programme to provide autologous and allogeneic hematopoietic stem cells transplant (HSCT) for adult and paediatric patients in the UAE. The programme is led by a team of top-notch hematologists and clinicians, offering advanced holistic care for patients with hematological diseases, in addition to genetic and autoimmune disorders. Since the launch of ADSCC in 2018, the centre has achieved various milestones such as manufacturing the first CAR-T Cell Therapy in UAE. It has also received multiple prestigious accreditations including its recognition as a Centre of Excellence in Hematopoietic Stem Cell Transplantation by the Department of Health - Abu Dhabi in 2023.
Prof. Yendry Ventura, CEO of ADSCC and Adjunct Professor at UAE University, said: Under the guidance of UAEs wise leadership and in line with the UAEs national agenda in healthcare, ADSCC has a clear vision and mandate of pioneering cellular therapy on a global scale and bringing medical breakthroughs closer to bedside, which plays a crucial role in positioning Abu Dhabi and the UAE at the vanguard of healthcare innovation. With this landmark FACT accreditation, we are breaking new ground in addressing complex diseases through cutting-edge treatments such as stem cell transplants and other cellular therapies. The journey towards the accreditation has been both extensive and stimulating, and we take immense pride in being the first institution in UAE to receive it. This achievement reaffirms our commitment to continue investing in providing the highest standards of patient care and offering top-tier cellular therapy services that is on par with the best globally.
Dr Phyllis Warkentin, FACT Chief Medical Officer, said: FACT accreditation represents a commitment to quality throughout an organisation and requires dedication and perseverance. I congratulate Prof. Ventura and the team of the Abu Dhabi Stem Cells Centre on achieving FACT accreditation, the first cellular therapy product processing laboratory in the UAE to reach this milestone.
The FACT accreditation programme conducts rigorous and comprehensive inspections awarding accreditation objectively based on evidence and in accordance with international standards and best practices in cellular therapies and is considered the leading accreditation body for cellular therapy programs worldwide.
Bone-marrow-homing lipid nanoparticles for genome editing in diseased and malignant haematopoietic stem cells – Nature.com
Laurenti, E. & Gottgens, B. From haematopoietic stem cells to complex differentiation landscapes. Nature 553, 418426 (2018).
Article CAS PubMed PubMed Central Google Scholar
Bauer, T. R. Jr. et al. Correction of the disease phenotype in canine leukocyte adhesion deficiency using ex vivo hematopoietic stem cell gene therapy. Blood 108, 33133320 (2006).
Article CAS PubMed PubMed Central Google Scholar
Blaese, R. M. et al. T lymphocyte-directed gene therapy for ADA-SCID: initial trial results after 4years. Science 270, 475480 (1995).
Article CAS PubMed Google Scholar
Boztug, K. et al. Stem-cell gene therapy for the Wiskott-Aldrich syndrome. N. Engl. J. Med. 363, 19181927 (2010).
Article CAS PubMed PubMed Central Google Scholar
Cowan, M. J. et al. Early outcome of a phase I/II clinical trial (NCT03538899) of gene-corrected autologous CD34+ hematopoietic cells and low-exposure busulfan in newly diagnosed patients with Artemis-deficient severe combined immunodeficiency (ART-SCID). Biol. Blood Marrow Transpl. 26, S88S89 (2020).
Article Google Scholar
Gaspar, H. B. et al. Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector. Lancet 364, 21812187 (2004).
Article CAS PubMed Google Scholar
Kanter, J. et al. Biologic and clinical efficacy of LentiGlobin for sickle cell disease. N. Engl. J. Med. 386, 617628 (2022).
Article CAS PubMed Google Scholar
Kohn, L. A. & Kohn, D. B. Gene therapies for primary immune deficiencies. Front. Immunol. 12, 648951 (2021).
Article CAS PubMed PubMed Central Google Scholar
Kondo, M. et al. Biology of hematopoietic stem cells and progenitors: implications for clinical application. Annu Rev. Immunol. 21, 759806 (2003).
Article CAS PubMed Google Scholar
Locatelli, F. et al. Betibeglogene autotemcel gene therapy for non-0/0 genotype -thalassemia. N. Engl. J. Med. 386, 415427 (2022).
Article CAS PubMed Google Scholar
Malech, H. L. et al. Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proc. Natl Acad. Sci. USA 94, 1213312138 (1997).
Article CAS PubMed PubMed Central Google Scholar
Morgan, R. A., Gray, D., Lomova, A. & Kohn, D. B. Hematopoietic stem cell gene therapy: progress and lessons learned. Cell Stem Cell 21, 574590 (2017).
Article CAS PubMed PubMed Central Google Scholar
Sago, C. D. et al. Nanoparticles that deliver RNA to bone marrow identified by in vivo directed evolution. J. Am. Chem. Soc. 140, 1709517105 (2018).
Article CAS PubMed PubMed Central Google Scholar
Shi, D., Toyonaga, S. & Anderson, D. G. In vivo RNA delivery to hematopoietic stem and progenitor cells via targeted lipid nanoparticles. Nano Lett. 23, 29382944 (2023).
Article CAS PubMed PubMed Central Google Scholar
Sou, K., Goins, B., Oyajobi, B. O., Travi, B. L. & Phillips, W. T. Bone marrow-targeted liposomal carriers. Expert Opin. Drug Deliv. 8, 317328 (2011).
Article CAS PubMed PubMed Central Google Scholar
Sou, K., Klipper, R., Goins, B., Tsuchida, E. & Phillips, W. T. Circulation kinetics and organ distribution of Hb-vesicles developed as a red blood cell substitute. J. Pharmacol. Exp. Ther. 312, 702709 (2005).
Article CAS PubMed Google Scholar
Xue, L. et al. Rational design of bisphosphonate lipid-like materials for mRNA delivery to the bone microenvironment. J. Am. Chem. Soc. 144, 99269937 (2022).
Article CAS PubMed Google Scholar
Boulais, P. E. & Frenette, P. S. Making sense of hematopoietic stem cell niches. Blood 125, 26212629 (2015).
Article CAS PubMed PubMed Central Google Scholar
Ikonomi, N., Kuhlwein, S. D., Schwab, J. D. & Kestler, H. A. Awakening the HSC: dynamic modeling of HSC maintenance unravels regulation of the TP53 pathway and quiescence. Front. Physiol. 11, 848 (2020).
Article PubMed PubMed Central Google Scholar
Li, J. Quiescence regulators for hematopoietic stem cell. Exp. Hematol. 39, 511520 (2011).
Article PubMed Google Scholar
Man, Y., Yao, X., Yang, T. & Wang, Y. Hematopoietic stem cell niche during homeostasis, malignancy, and bone marrow transplantation. Front. Cell Dev. Biol. 9, 621214 (2021).
Article PubMed PubMed Central Google Scholar
Nakamura-Ishizu, A., Takizawa, H. & Suda, T. The analysis, roles and regulation of quiescence in hematopoietic stem cells. Development 141, 46564666 (2014).
Article CAS PubMed Google Scholar
Eppert, K. et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat. Med. 17, 10861093 (2011).
Article CAS PubMed Google Scholar
Lapidot, T. et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 367, 645648 (1994).
Article CAS PubMed Google Scholar
Mandal, T., Beck, M., Kirsten, N., Linden, M. & Buske, C. Targeting murine leukemic stem cells by antibody functionalized mesoporous silica nanoparticles. Sci. Rep. 8, 989 (2018).
Article PubMed PubMed Central Google Scholar
Pei, S. & Jordan, C. T. How close are we to targeting the leukemia stem cell? Best Pract. Res. Clin. Haematol. 25, 415418 (2012).
Article CAS PubMed Google Scholar
Li, C. et al. Prophylactic in vivo hematopoietic stem cell gene therapy with an immune checkpoint inhibitor reverses tumor growth in syngeneic mouse tumor models. Cancer Res. 80, 549560 (2020).
Article CAS PubMed Google Scholar
Li, C. et al. In vivo HSPC gene therapy with base editors allows for efficient reactivation of fetal globin in beta-YAC mice. Blood Adv. 5, 11221135 (2021).
Article CAS PubMed PubMed Central Google Scholar
Li, C. et al. In vivo HSC gene therapy using a bi-modular HDAd5/35++ vector cures sickle cell disease in a mouse model. Mol. Ther. 29, 822837 (2021).
Article CAS PubMed Google Scholar
Li, C. et al. Safe and efficient in vivo hematopoietic stem cell transduction in nonhuman primates using HDAd5/35++ vectors. Mol. Ther. Methods Clin. Dev. 24, 127141 (2022).
Article PubMed Google Scholar
Psatha, N. et al. Enhanced HbF reactivation by multiplex mutagenesis of thalassemic CD34+ cells in vitro and in vivo. Blood 138, 15401553 (2021).
Article CAS PubMed PubMed Central Google Scholar
Muruve, D. A., Barnes, M. J., Stillman, I. E. & Libermann, T. A. Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Hum. Gene Ther. 10, 965976 (1999).
Article CAS PubMed Google Scholar
Sweeney, C. L. & De Ravin, S. S. The promise of in vivo HSC prime editing. Blood 141, 20392040 (2023).
Article CAS PubMed Google Scholar
Worgall, S., Wolff, G., Falck-Pedersen, E. & Crystal, R. G. Innate immune mechanisms dominate elimination of adenoviral vectors following in vivo administration. Hum. Gene Ther. 8, 3744 (1997).
Article CAS PubMed Google Scholar
Lek, A. et al. Death after high-dose rAAV9 gene therapy in a patient with Duchennes muscular dystrophy. N. Engl. J. Med. 389, 12031210 (2023).
Article CAS PubMed Google Scholar
Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 10781094 (2021).
Article CAS PubMed PubMed Central Google Scholar
Cheng, Q. et al. Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery and CRISPR-Cas gene editing. Nat. Nanotechnol. 15, 313320 (2020).
Article CAS PubMed PubMed Central Google Scholar
Dilliard, S. A., Cheng, Q. & Siegwart, D. J. On the mechanism of tissue-specific mRNA delivery by selective organ targeting nanoparticles. Proc. Natl Acad. Sci. USA 118, e2109256118 (2021).
Article CAS PubMed PubMed Central Google Scholar
Dilliard, S. A. & Siegwart, D. J. Passive, active and endogenous organ-targeted lipid and polymer nanoparticles for delivery of genetic drugs. Nat. Rev. Mater. 8, 282300 (2023).
Article CAS PubMed PubMed Central Google Scholar
Farbiak, L. et al. All-in-one dendrimer-based lipid nanoparticles enable precise HDR-mediated gene editing in vivo. Adv. Mater. 33, e2006619 (2021).
Article PubMed PubMed Central Google Scholar
Liu, S. et al. Membrane-destabilizing ionizable phospholipids for organ-selective mRNA delivery and CRISPR-Cas gene editing. Nat. Mater. 20, 701710 (2021).
Article CAS PubMed PubMed Central Google Scholar
Liu, S. et al. Zwitterionic phospholipidation of cationic polymers facilitates systemic mRNA delivery to spleen and lymph nodes. J. Am. Chem. Soc. 143, 2132121330 (2021).
Article CAS PubMed PubMed Central Google Scholar
Wang, X. et al. Preparation of selective organ-targeting (SORT) lipid nanoparticles (LNPs) using multiple technical methods for tissue-specific mRNA delivery. Nat. Protoc. 18, 265291 (2023).
Article CAS PubMed Google Scholar
Wei, T., Cheng, Q., Min, Y. L., Olson, E. N. & Siegwart, D. J. Systemic nanoparticle delivery of CRISPR-Cas9 ribonucleoproteins for effective tissue specific genome editing. Nat. Commun. 11, 3232 (2020).
Article CAS PubMed PubMed Central Google Scholar
Zhang, D. et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nat. Nanotechnol. 17, 777787 (2022).
Article CAS PubMed PubMed Central Google Scholar
Wu, L. C. et al. Correction of sickle cell disease by homologous recombination in embryonic stem cells. Blood 108, 11831188 (2006).
Article CAS PubMed PubMed Central Google Scholar
Metais, J. Y. et al. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv. 3, 33793392 (2019).
Article PubMed PubMed Central Google Scholar
Newby, G. A. et al. Base editing of haematopoietic stem cells rescues sickle cell disease in mice. Nature 595, 295302 (2021).
Article CAS PubMed PubMed Central Google Scholar
Stavropoulou, V., Peters, A. & Schwaller, J. Aggressive leukemia driven by MLL-AF9. Mol. Cell Oncol. 5, e1241854 (2018).
Article PubMed Google Scholar
Hou, X. et al. Vitamin lipid nanoparticles enable adoptive macrophage transfer for the treatment of multidrug-resistant bacterial sepsis. Nat. Nanotechnol. 15, 4146 (2020).
Article CAS PubMed PubMed Central Google Scholar
The rest is here:
Bone-marrow-homing lipid nanoparticles for genome editing in diseased and malignant haematopoietic stem cells - Nature.com
COVID-19 was associated with the complications after allogeneic hematopoietic stem cell transplantation | Scientific … – Nature.com
Patients characteristics
The characteristics of consecutive 179 hospitalized COVID-19 patients were showed in Table 1. The comparison between serious and non-serious infection group was showed in Supplementary Table 1. Forty-three (24%) and 136 (76%) patients were diagnosed as serious and non-serious infection, respectively. The median duration of disease was 18days (range 2102) days, and duration of SARS-CoV-2 infection>18days and18days was defined as long-term (n=90) and short-term infection (n=89), respectively (Supplementary Table 2). The median time from transplantation to COVID-19 infection occurrence was 149days (range 63038) days. A total of 116 (64.8%) patients received immunosuppressants, including cyclosporin (n=85), tacrolimus (n=8), ruxolitinib (n=8), or glucocorticoids (n=32) when COVID-19 occurrence. Seventeen patients received more than 1 type of immunosuppressants. A total of 144 (80.4%) patients received anti-viral treatment (140 for Paxlovid and 4 for Azvudine). Forty serious infection cases (93.0%) received anti-viral treatment (39 for Paxlovid and 1 for Azifudine), 85 long-term infection cases (94.4%) received anti-viral treatment (83 for Paxlovid and 2 for Azifudine). Sixty-two patients received newly added drugs corticosteroid treatment, that is, 23 (53.5%) and 39 (28.7%) patients were in the serious and non-serious infection group, 9 (10.1%) and 53 (58.9%) patients were in the short- and long-term infection group, respectively.
A total of 26, 33, and 11 patients showed PGF, leukopenia and thrombocytopenia, respectively, after COVID 19 infection. The 150-day cumulative incidence of leukopenia, thrombocytopenia, and PGF was 18.4% (95% confidence interval [CI] 12.724.1%), 6.1% (95% CI 2.619.6%), and 14.5% (95% CI 9.319.7%) respectively, after COVID-19.
The 150-day cumulative incidence of PGF after COVID-19 was 11.8% (95% CI 6.417.2%) versus 23.3% (95% CI 10.536.1%) (P=0.071) between non-serious and serious infection group, which was 9.0% (95% CI 3.015.0%) versus 20.0% (95% CI 11.728.3%) (P=0.045) between short- and long-term infection group (Fig.1A).
The 150-day cumulative incidence of PGF and leukopenia after COVID-19 between short and long-term infection group. (A) PGF; (B) leukopenia. PGF poor graft function.
The 150-day cumulative incidence of leukopenia after COVID-19 was 19.9% (95% CI 13.226.6%) versus 14.0% (95% CI 3.524.5%) (P=0.325) between non-serious and serious infection group, which was 12.4% (95% CI 5.519.3%) versus 24.4% (95% CI 15.533.3%) (P=0.049) between short- and long-term infection group (Fig.1B).
The 150-day cumulative incidence of thrombocytopenia after COVID-19 was 5.1% (95% CI 1.48.8%) versus 9.3% (95% CI 0.518.1%) (P=0.331) between non-serious and serious infection group, which was 5.6% (95% CI 0.810.4%) versus 6.7% (95% CI 1.511.9%) between short- and long-term infection group (P=0.779).
The median duration of leukopenia, thrombocytopenia, and PGF was 14days (range 4118) days, 10days (range 878) days, and 17days (range 978) days, respectively. Until the last follow up, leukopenia, thrombocytopenia and PGF were still persistence 22 patients.
In multivariable analysis, after adjusted by other factors, the duration of COVID-19 was associated with PGF (hazard ratio [HR], 2.31; 95% CI 1.045.11; P=0.039) and leukopenia (HR 2.29; 95% CI 1.045.07; P=0.04) (Table 2). The other risk factors for PGF and leukopenia were showed in Supplementary Table 4.
A total of 5 patients showed aGVHD after COVID 19 infection, and the cumulative incidence of total aGVHD was 2.8% (95% CI 0.45.2%) after COVID-19. The cumulative incidence of aGVHD after COVID-19 was 3.7% (95% CI 0.56.9%) versus 0% (P=0.204), respectively, between non-serious and serious infection group, which was 3.4% (95% CI 0.47.2%) versus 2.2% (95% CI 0.95.3%) (P=0.651), respectively, between short- and long-term infection group. No risk factors were associated with aGVHD in multivariable analysis.
A total of 4, 4 and 3 patients showed mild, moderate, and severe cGVHD after COVID 19 infection, and the cumulative incidence of cGVHD was 6.70% after COVID-19. The 150-day cumulative incidence of cGVHD after COVID-19 was 7.4% (95% CI 2.911.8%) versus 4.7% (95% CI 1.711.1%), respectively, between non-serious and serious infection group (P=0.544). The 150-day cumulative incidence of cGVHD after COVID-19 was 6.7% (95% CI 1.511.9%) versus 5.6% (95% CI 0.810.4%), respectively, between short- and long-term infection group (P=0.741). No risk factors were associated with cGVHD in multivariable analysis.
A total of 34 and 7 patients showed CMV DNAemia and CMV disease (CMV pneumonia: 5, CMV gastrointestinal disease: 1, CMV encephalitis+retinitis: 1) after COVID 19 infection. The 150-day cumulative incidence of CMV DNAemia after COVID-19 was 19.9% (95% CI 13.226.6%) versus 11.6% (95% CI 1.921.3%) (P=0.204), respectively, between non-serious and serious infection group, which was 14.6% (95% CI 7.222.0%) versus 23.3% (95% CI 14.532.1%) (P=0.118), respectively, between short- and long-term infection group. The 150-day cumulative incidence of CMV disease after COVID-19 was 0.7% (95% CI 0.72.1%) versus 14.0% (95% CI 3.524.5%) (P<0.0001, Fig.2A), respectively, between non-serious and serious infection group, which was 0% versus 7.8% (95% CI 2.213.4%) (P=0.007, Fig.2B), respectively, between short- and long-term infection group. Particularly, the 150-day cumulative incidence of CMV pneumonia after COVID-19 was 0% versus 11.6% (95% CI 1.921.4%) (P<0.0001, Fig.2C), respectively, between non-serious and serious infection group, which was 0% versus 5.6% (95% CI 0.810.4%) (P=0.0245, Fig.2D), respectively, between short- and long-term infection group. In multivariable analysis, after adjusted by other factors, the severity of COVID-19 was associated with CMV disease (HR, 20.15; 95% CI 2.43167.36, P=0.005) (Table 2).
The association between COVID-19 and CMV disease. The 150-day cumulative incidence of CMV disease after COVID-19 between (A) non-serious and serious infection group; (B) short and long-term infection group. The 150-day cumulative incidence of CMV pneumonia after COVID-19 between (C) non-serious and serious infection group; (D) short and long-term infection group. CMV cytomegalovirus.
A total of 11 and 3 patients showed Epstein-Barr virus (EBV) DNAemia and EBV associated posttransplant lymphoproliferative disorders (PTLD) after COVID-19. The 150-day cumulative incidence of EBV DNAemia after COVID-19 was 7.4% (95% CI 3.011.8%) versus 0.00% (P=0.068), respectively, between non-serious and serious infection group. The 150-day cumulative incidence of EBV DNAemia after COVID-19 was 5.6% (95% CI 0.810.4%) versus 5.6% (95% CI 0.810.4%) (P=0.968), respectively, between short- and long-term infection group. All the 3 PTLD patients were in the non-serious group. No risk factors were associated with EBV DNAemia and PTLD in multivariable analysis.
A total of 27 patients died after COVID-19, and the caused were summarized in Table 3. The most common cause was infection besides of COVID-19 (n=9, 33.3%), followed by relapse (n=7, 25.9%) and COVID-19 (n=4, 14.8%).
The 150-day cumulative incidence of non-relapse mortality (NRM) after COVID-19 infection was 11.2% (95% CI 6.615.8%), which was 2.2% (95% CI 0.34.7%) and 39.5% (95% CI 24.654.4%) between non-serious and serious infection group (P<0.0001, Fig.3A), and was 2.2% (95% CI 0.95.3%) and 20.0% (95% CI 11.728.3%) (P=0.002, Fig.3B) between short- and long-term infection group.
The association between COVID-19 and survival. The 150-day cumulative incidence of NRM after COVID-19 between (A) non-serious and serious infection group; (B) short and long-term infection group. The 150-day probability of OS after COVID-19 infection between (C) non-serious and serious infection group; (D) short and long-term infection group. NRM non-relapse mortality, OS overall survival.
The 150-day probability of overall survival (OS) after COVID-19 infection was 84.9% (95% CI 79.690.2%), which was 94.9% (95% CI 91.298.6%) and 53.5% (95% CI 38.168.5%) between non-serious and serious infection group (P<0.0001, Fig.3C), and was 93.3% (95% CI 88.198.5%) and 76.7% (95% CI 67.985.5%) (P=0.002, Fig.3D) between short- and long-term infection group.
In multivariable analysis, after adjusted by other factors, the severity of COVID-19 was associated with NRM (HR, 17.26; 95% CI 4.8761.21, P<0.0001). The severity of COVID-19 were associated with OS (HR, 14.00; 95% CI 5.8733.42, P<0.0001) (Table 2), The other risk factors for NRM and OS were showed in Supplementary Table 4.
A total of 179 patients without COVID-19 infection were enrolled as controlled and the characteristics between patients with and without COVID-19 were showed in Supplementary Table 5. The 150-day probability of NRM and OS were 11.2% (95% CI 6.615.8%) versus 3.9% (95% CI 1.16.7%) with P=0.009 and 84.9% (95% CI 79.690.2%) versus 93.9% (95% CI: 88.7%99.1%) with P=0.006, respectively, for patients in the group with and without COVID-19 infection (Fig.4A,B). The probability of NRM and OS for patients without COVID-19 infections were superior to those in serious infection group or long-term infection group (Fig.4C,D).
Clinical outcomes of patients with and without COVID 19 infection. (A) The 150-day cumulative incidence of NRM in the group with and without COVID-19 infection; (B) The 150-day cumulative incidence of OS in the group with and without COVID-19 infection; (C) The 150-day cumulative incidence of NRM in the group without COVID-19 infection, serious infection and long-term infection; (D) The 150-day cumulative incidence of OS in the group without COVID-19 infection, serious infection and long-term infection, NRM non-relapse mortality, OS overall survival.
Follow this link:
COVID-19 was associated with the complications after allogeneic hematopoietic stem cell transplantation | Scientific ... - Nature.com
Azacitidine and gemtuzumab ozogamicin as post-transplant maintenance therapy for high-risk hematologic … – Nature.com
Pavletic SZ, Kumar S, Mohty M, de Lima M, Foran JM, Pasquini M, et al. NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on the epidemiology and natural history of relapse following allogeneic cell transplantation. Biol Blood Marrow Transpl. 2010;16:87190.
Article Google Scholar
Porter DL, Alyea EP, Antin JH, DeLima M, Estey E, Falkenburg JH, et al. NCI first international workshop on the biology, prevention, and treatment of relapse after allogeneic hematopoietic stem cell transplantation: report from the committee on treatment of relapse after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transpl. 2010;16:1467503.
Article Google Scholar
Warlick ED, Cioc A, Defor T, Dolan M, Weisdorf D. Allogeneic stem cell transplantation for adults with myelodysplastic syndromes: importance of pretransplant disease burden. Biol Blood Marrow Transpl. 2009;15:308.
Article Google Scholar
Armistead PM, de Lima M, Pierce S, Qiao W, Wang X, Thall PF, et al. Quantifying the survival benefit for allogeneic hematopoietic stem cell transplantation in relapsed acute myelogenous leukemia. Biol Blood Marrow Transpl. 2009;15:14318.
Article Google Scholar
Kaito S, Wada A, Adachi H, Konuma R, Kishida Y, Nagata A, et al. Geriatric nutritional risk index as a useful prognostic factor in second allogeneic hematopoietic stem cell transplantation. Ann Hematol. 2020;99:165565.
Article CAS PubMed Google Scholar
Dombret H, Seymour JF, Butrym A, Wierzbowska A, Selleslag D, Jang JH, et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood. 2015;126:291-9.
Article CAS PubMed PubMed Central Google Scholar
Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009;10:22332.
Article CAS PubMed PubMed Central Google Scholar
Campoli M, Ferrone S. HLA antigen changes in malignant cells: epigenetic mechanisms and biologic significance. Oncogene. 2008;27:586985.
Article CAS PubMed PubMed Central Google Scholar
Goodyear O, Agathanggelou A, Novitzky-Basso I, Siddique S, McSkeane T, Ryan G, et al. Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood. 2010;116:190818.
Article CAS PubMed Google Scholar
Goodyear OC, Dennis M, Jilani NY, Loke J, Siddique S, Ryan G, et al. Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood. 2012;119:33619.
Article CAS PubMed Google Scholar
de Lima M, Giralt S, Thall PF, de Padua Silva L, Jones RB, Komanduri K, et al. Maintenance therapy with low-dose azacitidine after allogeneic hematopoietic stem cell transplantation for recurrent acute myelogenous leukemia or myelodysplastic syndrome: a dose and schedule finding study. Cancer. 2010;116:542031.
Article PubMed Google Scholar
Pusic I, Choi J, Fiala MA, Gao F, Holt M, Cashen AF, et al. Maintenance therapy with decitabine after allogeneic stem cell transplantation for acute myelogenous leukemia and myelodysplastic syndrome. Biol Blood Marrow Transpl. 2015;21:17619.
Article CAS Google Scholar
Craddock C, Jilani N, Siddique S, Yap C, Khan J, Nagra S, et al. Tolerability and clinical activity of post-transplantation azacitidine in patients allografted for acute myeloid leukemia treated on the RICAZA Trial. Biol Blood Marrow Transpl. 2016;22:38590.
Article CAS Google Scholar
El-Cheikh J, Massoud R, Fares E, Kreidieh N, Mahfouz R, Charafeddine M, et al. Low-dose 5-azacytidine as preventive therapy for relapse of AML and MDS following allogeneic HCT. Bone Marrow Transpl. 2017;52:91821.
Article CAS Google Scholar
de Lima M, Oran B, Champlin RE, Papadopoulos EB, Giralt SA, Scott BL, et al. CC-486 maintenance after stem cell transplantation in patients with acute myeloid leukemia or myelodysplastic syndromes. Biol Blood Marrow Transpl. 2018;24:201724.
Peiper SC, Ashmun RA, Look AT. Molecular cloning, expression, and chromosomal localization of a human gene encoding the CD33 myeloid differentiation antigen. Blood. 1988;72:31421.
Article CAS PubMed Google Scholar
Nand S, Othus M, Godwin JE, Willman CL, Norwood TH, Howard DS, et al. A phase 2 trial of azacitidine and gemtuzumab ozogamicin therapy in older patients with acute myeloid leukemia. Blood. 2013;122:34329.
Article CAS PubMed PubMed Central Google Scholar
Taksin AL, Legrand O, Raffoux E, de Revel T, Thomas X, Contentin N, et al. High efficacy and safety profile of fractionated doses of Mylotarg as induction therapy in patients with relapsed acute myeloblastic leukemia: a prospective study of the alfa group. Leukemia. 2007;21:6671.
Article CAS PubMed Google Scholar
Larson RA, Sievers EL, Stadtmauer EA, Lwenberg B, Estey EH, Dombret H, et al. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer. 2005;104:144252.
Article CAS PubMed Google Scholar
Oshikawa G, Kakihana K, Saito M, Aoki J, Najima Y, Kobayashi T, et al. Post-transplant maintenance therapy with azacitidine and gemtuzumab ozogamicin for high-risk acute myeloid leukaemia. Br J Haematol. 2015;169:7569.
Article CAS PubMed Google Scholar
de Greef GE, van Putten WL, Boogaerts M, Huijgens PC, Verdonck LF, Vellenga E, et al. Criteria for defining a complete remission in acute myeloid leukaemia revisited. An analysis of patients treated in HOVON-SAKK co-operative group studies. Br J Haematol. 2005;128:18491.
Article PubMed Google Scholar
Armand P, Kim HT, Zhang MJ, Perez WS, Dal Cin PS, Klumpp TR, et al. Classifying cytogenetics in patients with acute myelogenous leukemia in complete remission undergoing allogeneic transplantation: a Center for International Blood and Marrow Transplant Research study. Biol Blood Marrow Transpl. 2012;18:2808.
Article Google Scholar
Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:42447.
Article PubMed PubMed Central Google Scholar
Armand P, Kim HT, Logan BR, Wang Z, Alyea EP, Kalaycio ME, et al. Validation and refinement of the disease risk index for allogeneic stem cell transplantation. Blood. 2014;123:366471.
Article CAS PubMed PubMed Central Google Scholar
Najima Y, Sadato D, Harada Y, Oboki K, Hirama C, Toya T, et al. Prognostic impact of TP53 mutation, monosomal karyotype, and prior myeloid disorder in nonremission acute myeloid leukemia at allo-HSCT. Bone Marrow Transplant. 2021;56:33446.
Article CAS PubMed Google Scholar
Ikegame K, Yoshida T, Yoshihara S, Daimon T, Shimizu H, Maeda Y, et al. Unmanipulated haploidentical reduced-intensity stem cell transplantation using fludarabine, busulfan, low-dose antithymocyte globulin, and steroids for patients in non-complete remission or at high risk of relapse: a prospective multicenter phase I/II study in Japan. Biol Blood Marrow Transplant. 2015;21:1495505.
Article CAS PubMed Google Scholar
Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, et al. 1994 consensus conference on acute GVHD grading. Bone Marrow Transplant. 1995;15:8258.
CAS PubMed Google Scholar
Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, et al. National institutes of health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transpl. 2015;21:389401.e1.
Article Google Scholar
Kanda Y. Investigation of the freely available easy-to-use software EZR for medical statistics. Bone Marrow Transpl. 2013;48:4528.
Article CAS Google Scholar
de Lima M. New approaches to transplantation in acute myelogenous leukemia. Hematology Am Soc Hematol Educ Program. 2015;2015:596604.
Article PubMed Google Scholar
Rajvanshi P, Shulman HM, Sievers EL, McDonald GB. Hepatic sinusoidal obstruction after gemtuzumab ozogamicin (Mylotarg) therapy. Blood. 2002;99:23104.
Article CAS PubMed Google Scholar
McKoy JM, Angelotta C, Bennett CL, Tallman MS, Wadleigh M, Evens AM, et al. Gemtuzumab ozogamicin-associated sinusoidal obstructive syndrome (SOS): an overview from the research on adverse drug events and reports (RADAR) project. Leuk Res. 2007;31:599604.
Article CAS PubMed Google Scholar
Oran B, de Lima M, Garcia-Manero G, Thall PF, Lin R, Popat U, et al. A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Adv. 2020;4:55808.
Article CAS PubMed PubMed Central Google Scholar
Hassan MA, Moukalled N, El Cheikh J, Bazarbachi A, Abou Dalle I. Azacitidine in combination with venetoclax maintenance post-allogeneic hematopoietic stem cell transplantation in T cell acute lymphoblastic leukemia. Clin Hematol Int. 2023;5:525.
Article PubMed PubMed Central Google Scholar
El-Cheikh J, Bidaoui G, Saleh M, Moukalled N, Abou Dalle I, Bazarbachi A. Venetoclax: a new partner in the novel treatment era for acute myeloid leukemia and myelodysplastic syndrome. Clin Hematol Int. 2023;5:14354.
Article PubMed PubMed Central Google Scholar
Levis M, Hamadani M, Logan B, Jones R, Singh A, Litzow M, et al. Gilteritinib as post-transplant maintenance for acute myeloid leukemia with internal tandem duplication mutation of FLT3. J Clin Oncol. 2024:JCO2302474. https://doi.org/10.1200/JCO.23.02474. Online ahead of print.
Platzbecker U, Wermke M, Radke J, Oelschlaegel U, Seltmann F, Kiani A, et al. Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia. 2012;26:3819.
Article CAS PubMed Google Scholar
Schroeder T, Rachlis E, Bug G, Stelljes M, Klein S, Steckel NK, et al. Treatment of acute myeloid leukemia or myelodysplastic syndrome relapse after allogeneic stem cell transplantation with azacitidine and donor lymphocyte infusions-a retrospective multicenter analysis from the German Cooperative Transplant Study Group. Biol Blood Marrow Transpl. 2015;21:65360.
Article CAS Google Scholar
Selective haematological cancer eradication with preserved haematopoiesis – Nature.com
Structural dataset and computational analysis
The experimentally determined 3D structure of the CD45 extracellular domain was retrieved from the PDB (5FMV)22. The per-residue relative solvent accessibility area was computed using a previously published algorithm45 implemented in FreeSASA46 using default parameters. Prediction of B-cell epitopes was based on BepiPred-2.0 (ref. 47) using the default threshold (0.5) for epitope residues nomination. The EV mutation sequence analysis framework48 was used to search for the CD45 sequence with the non-redundant UniProtKB database49. A multiple-sequence alignment was built using five iterations of the jackhammer HMM search algorithm50 with default significance score for the inclusion of homologous sequences.
For sgRNA cloning into the px458 host vector (a gift from F. Zhang) (Supplementary Table 1), forward and reverse primers containing complementary CRISPR RNA (crRNA) sequences flanked by BbsI restriction sites were used (Supplementary Table 4). The px458 plasmid was double digested with AgeI-HF (NEB, R3552S) and EcoRI-HF (NEB, R3101S) to eliminate the regions coding for GFP and Cas9. The px458 vector was then digested using BbsI (ThermoFisher Scientific, ER1012), gel purified and ligated with the phosphorylated and annealed crRNA oligonucleotides (called sgRNA plasmid once cloned).
To transiently overexpress CD45 mutants, we introduced each variant of interest in a plasmid expressing WT CD45RO. Briefly, we digested the pCD45RABC plasmid (Sino Biological) (Supplementary Table 1) with HindIII-HF (NEB, R3104S) and XcmI (NEB, R0533L) to remove the alternative spliced exons A, B and C. The point mutations of the human CD45 variants were then introduced into the plasmid expressing CD45RO using PCR (Supplementary Table 4).
All ligations were transformed in JM109-competent bacteria (Promega, P9751). BE plasmids were from Addgene (SPACE-NG, ABE8e-NG, ABEmax-SpRY, ABEmax-SpG, CBE4max-NG, CBE4max-SpG and xCas9(3.7)-BE4) (Supplementary Table 1).
ABE8e-NG mRNA (capped (cap 1) using CleanCap AG; fully substituted with 5-methoxy-U; 120A polyA tail) and ABE8e(TadA-8e V106W)-SpRY mRNA (capped (cap 1) using CleanCap AG 3-O-methylation; fully substituted with N1-methyl-pseudo-U; 80A polyA tail) were from Trilink Biotechnologies and Tebu-bio. We used 100-base lyophilized chemically modified sgRNAs from Synthego using their CRISPRevolution sgRNA EZ Kit service and resuspended at 100M (3.2gl1) in 1 TE buffer from Synthego (10nM Tris, 1mM EDTA, pH 8.0; chemical modifications include 2-O-methylation of the three first and last bases and 3 phosphorothioate bonds between the first three and last two bases of each sgRNA).
Cells from the BE plasmid screening were lysed in tail lysis buffer (100mM Tris pH8.5, 5mM Na-EDTA, 0.2%SDS, 200mM NaCl) containing proteinase K (Sigma-Aldrich) at 56C (1,000rpm) for 1h. The DNA was precipitated with isopropanol (1:1 volume ratio) and washed in 70% ethanol. The DNA was then resuspended in H2O and the genomic DNA concentration was measured with a NanoDrop device (Thermo Fisher).
For samples containing few cells, genomic DNA was extracted using QuickExtract (Lucigen, QE09050). Cell pellets were resuspended in 30l QuickExtract, incubated at 60C for 6min, vortexed for 1min and subsequently re-incubated at 98C for 10min.
PCR was performed using GoTaq G2 Green Master Mix (Promega, M782B). The gDNA of samples analysed by NGS was extracted using QuickExtract (Lucigen, QE09050) or the Quick-DNA 96 Plus kit (Zymo, D4070) and the genomic DNA concentration was measured with a Qubit device (Thermo Fisher).
For Sanger sequencing, different PCR primers were used depending on the CD45 exon targeted by the sgRNA and the sequencing technology (Supplementary Table 4). Sequencing of PCR amplicons was done at Microsynth and sequencing chromatograms were analysed using the EditR R package51 to quantify BE efficiencies.
For NGS, targeted amplicon libraries were generated using a three-step PCR protocol. In brief, nested PCRs were done on genomic DNA samples using KAPA HiFi HotStart polymerase (Roche) (Supplementary Table 4). After Illumina barcoding (Nextera indices, Illumina) using KAPA HiFi HotStart polymerase (Roche), PCR samples were pooled, purified using AMPure XP beads (Beckman Coulter) and quantified using Qubit dsDNA HS assay kit (Thermo Fisher). Libraries were paired-end sequenced on an Illumina Miniseq instrument using the Illumina Miniseq Mid output kit (300 cycles) with 50% PhiX spike-in (Illumina). After demultiplexing, each sample was assessed for quality using FastQC52 and processed using the CRISPResso2 tool53. For each of the samples, we provided the reference amplicon sequence (hg38) and the guide RNA sequence (reverse complement) and defined the quantification window centre to 10, the quantification window size to 15 and the plot window size to 30. We applied minimum paired end reads overlap between 10 and 200 and provided the following Trimmomatic sentence: ILLUMINACLIP:NexteraPE-PE:2:30:10 LEADING:3 TRAILING:3 SLIDINGWINDOW:4:15 MINLEN:36. Finally, we used a custom R script (https://gitlab.com/JekerLab/cd45_shielding) to count and translate into amino acid each allele from the CRISPResso2 output file Alleles_frequency_table.txt in the quantification window. Alleles with less than 0.8% frequency were considered as other.
Genetic discrimination between PDX- and HSPC-derived cells was performed using the Devyser Chimerism NGS kit (Devyser) according to the manufacturers recommendations. In brief, sequencing libraries were prepared from genomic DNA targeting 24 polymorphic insertiondeletion markers distributed on 16 different chromosomes. The libraries were sequenced on a MiniSeq (Illumina) instrument using the high-throughput flow cell, generating 74-bp pair-end reads. An informative marker set was defined for the PDX donor/HSPC donor pair. It consisted of 10 markers that reliably discriminated between DNA from PDX and HSPC donor cells. The average proportion of leukaemia donor-specific reads to total reads was calculated to determine the proportion of PDX cells in each sample. It was confirmed that the genomic DNA of the mouse host did not interfere with the analysis.
Genomic DNA was extracted from human peripheral blood mononuclear cells (PBMCs) using the Puregene tissue kit (Qiagen, 158063) according to the manufacturers instructions including the proteinase-K and RNase steps (Qiagen, 158143 and 158153). CHANGE-seq-BE was adapted and modified from the original CHANGE-seq method54 to validate genome-wide activity for ABEs28. Similar to CHANGE-seq, purified genomic DNA tagmented with a custom Tn5-transposome to generate an average length of 650bp and followed by gap repair with Kapa HiFi HotStart Uracil + DNA Polymerase (KAPA Biosystems, KK2802) and Taq DNA ligase (NEB, M0208L). Gap-repaired DNA was treated with USER enzyme (NEB, M5505L) and T4 polynucleotide kinase (NEB, M0201L). Intramolecular circularization of the DNA was performed with T4 DNA ligase (NEB, M0202L) and residual linear DNA was degraded by a cocktail of exonucleases containing plasmid-safe ATP-dependent DNase (Lucigen, E3110K), lambda exonuclease (NEB, M0262L), exonuclease I (NEB, M0293L) and exonuclease III (NEB, M0206L). The circularized DNA was then treated with Quick CIP (NEB, M025L) to dephosphorylate 5 and 3 ends of any residual linear DNA. Circularized genomic DNA (125ng) was treated with ABE8eSpRY:sgRNA-49.3 complexes in vitro in a 50l reaction for 24h at 37C. ABE RNP complexes nicked the targeted DNA strand and deaminated adenine bases to inosine in the non-targeted stranded DNA of both on- and off-target sites. Further enzymatic steps were included with ABE treatment in the CHANGE-seq-BE method to generate double-strand breaks. Nicked DNA circles were treated with endonuclease V in 10 NEB buffer 4 (NEB, M0305S). Endo V cleaved DNA adjacent to inosines to generate linear DNA with 5 overhangs. Gaps were filled with klenow fragments (3>5 exo) and deoxyribonucleotide triphosphates (dNTPs) (NEB, M0212L) in NEB buffer 2. End-repaired DNA products were A-tailed and further ligated with a hairpin adapter using an HTP library preparation kit (Kapa, KK8235), USER treated and amplified by PCR-barcoded universal primers with NEBNext multiplex oligonucleotides for illumina (NEB, E7600S), using Kapa HiFi HotStart uracil master mix. PCR libraries were quantified by quantitative PCR (KAPA Biosystems, KK4824) and sequenced with 151-8-8151 cycles on an Illumina NextSeq 2000 instrument. CHANGE-seq-BE data analyses were performed using open-source software: https://github.com/tsailabSJ/changeseq/tree/dev.
Validation of off-target sites was performed using the rhAmpSeq system from IDT. rhAmpSeq primer panels for targeted amplification were generated using the rhAmpSeq design tool defining the insert size between 150 and 250bp. Applied primer sequences are listed in Supplementary Table 4. A rhAmpSeq CRISPR library was prepared according to the manufacturers instructions and sequenced on an Illumina MiniSeq instrument (MiniSeq high output kit, 300 cycles). Custom python code and open-source bioinformatic tools were used to analyse rhAmpSeq data. First, we generated FASTQ format files by demultiplexing high-throughput-sequencing BCL data files. Next, the reads were processed using CRISPRessoPooled (v.2.0.41) with quantification_window_size 10, quantification_window_center 10, base_editor_output, conversion_nuc_from A, conversion_nuc_to G. The allele frequency table from the output files was used to calculate the AT-to-GC editing frequency. Specifically, the editing frequency for each on- or off-target site was defined as the ratio between the number of reads containing the edited base (that is, G) in a window from positions 4 to 10 of each protospacer (where the GAA PAM is positions 2123) and the total number of reads. To calculate the statistical significance of off-target editing, we applied a method previously described55. In brief, a 2-by-2 contingency table was constructed using the number of edited reads and the number of unedited reads in the treated sample and its corresponding control sample. Next, a 2 test was done. The FDR was calculated using the BenjaminiHochberg method. Significant off-targets were defined on the basis of: first, FDR0.05 and second, the difference in editing frequency between treated and control (1%).
All cancer cell lines (listed in Supplementary Table 5) were cultured in RPMI-1640 (Sigma-Aldrich, R8758) supplemented with 10% heat-inactivated FCS (Gibco Life Technologies) and 2mM GlutaMAX (ThermoFisher Scientific, 35050061) at 37C.
All cell lines were retrovirally transduced with MI-Luciferase-IRES-mCherry (gift from X. Sun; Supplementary Table 1). Cells were then FACS-sorted on the basis of mCherry expression. After expansion, MOLM-14 and OCIAML2 were profiled for short tandem repeats and tested negative for mycoplasma before being frozen until further use. Jurkat and NALM-6 were purchased from ATCC and were therefore not profiled for short tandem repeats.
DF-1 cells were cultured in DMEM high-glucose medium (Sigma-Aldrich, D5796) supplemented with 10% non-heat-inactivated FCS and 2mM GlutaMAX at 39C (Supplementary Table 5).
Leukocyte buffy coats from anonymous healthy human donors were purchased from the blood-donation centre at Basel (Blutspendezentrum SRK beider Basel, BSZ). PBMCs were isolated by density centrifugation using SepMate tubes (StemCell Technologies, 85450) and the density gradient medium Ficoll-Paque (GE Healthcare) according to the manufacturers protocol. Frozen PBMCs were thawed and human primary T cells were then isolated using an EasySep human T cell isolation kit (Stemcell Technologies, 17951) following the manufacturers protocol. T cells were cultured overnight without stimulation at a density of 1.5106 cells per ml in RPMI-1640 medium supplemented with 10% heat-inactivated human serum (AB+, male; purchased from BSZ), 10mM HEPES (Sigma-Aldrich), 2mM GlutaMAX, 1mM sodium pyruvate, 0.05mM 2-mercaptoethanol and 1% MEM non-essential amino acids (all from Gibco Life Technologies). The next day, the human primary T cells were activated with interleukin-2 (IL-2) (150Uml1, proleukin, University Hospital Basel), IL-7 (5ngml1, R&D Systems), IL-15 (5ngm1, R&D Systems) and Dynabead Human T-Activator CD3/CD28 (1:1 beads:cells ratio) (Gibco, 11132D). The activated cells were de-beaded before electroporation.
Leukopaks were purchased from CytoCare and hCD34+ HSPCs were isolated by the LP-34 process using CliniMACS Prodigy (Miltenyi). Isolated hCD34+ HSPCs were thawed and grown in HSPC medium for two days until electroporation (StemSpan SFEM II (StemCell, 09655) supplemented with 100ngml1 human stem cell factor (hSCF) (Miltenyi, 130-096-695), 100ngml1 human FMS-like tyrosine kinase ligand (hFlt3)-ligand (Miltenyi, 130-096-479), 100ngml1 human thrombopoietin (hTPO) (Miltenyi, 130-095-752) and 60ngml1 hIL-3 (Miltenyi, 130-095-069).
K562 cells (2106) were resuspended in buffer T and mixed with 5g BE plasmid (Supplementary Table 1) and 1.5g sgRNA plasmid for co-electroporation using a Neon transfection system (ThermoFisher, MPK10096; 1,450V, 10ms, 3 pulses). To monitor the electroporation efficiency, GFP expression was evaluated 24h after electroporation using an optical microscope. In Extended Data Fig. 1f, BE results are displayed using a custom BE score: log((sum of editing frequencies per condition/number of edited positions per condition)+1).
De-beaded human activated T cells (1106) were resuspended in 100l Lonza supplemented P3 electroporation buffer with 7.5g BE mRNA (1gl1) and 7.5g sgRNA (3.2gl1) (Supplementary Table 2) and electroporated using the 4D-Nucleofector system (Lonza) with program EH-115. Immediately after electroporation, 900l pre-warmed human T-cells medium was added directly in the cuvettes and incubated for 20min at 37C for the T cells to recover. Cells were then transferred in 48-well flat-bottomed plates (Corning, 3548) and the medium was supplemented with 500Uml1 IL-2. The medium was renewed every two days.
hCD34+ HSPCs (1106) were electroporated 48h after thawing with 7.5g BE mRNA (1gl1) and 13.6g sgRNA (3.2gl1) (Supplementary Table 2) with a 1:100 BE:sgRNA molar ratio, following the same protocol as for human T cells but with program CA-137. Electroporated hCD34+ HSPCs were kept in culture at 0.5106 cells per ml in a six-well flat-bottomed plate (Corning, 3516) in HSPC medium supplemented with 100ngml1 hSCF, 100ngml1 hFlt3ligand and 100ngml1 hTPO for in vivo applications and with the addition of 60ngml1 hIL-3 for in vitro assays. The medium was renewed every five days. Edited hCD34+ HSPCs prepared for in vivo injection were frozen two days after electroporation in cryo-preservation CryoStor CS10 medium (Stem Cell Technologies, 07930) at a density of 10106 cells ml1.
The CFU assay was started 72h after gene editing. For each condition, 1.1ml semi-solid methylcellulose medium (StemCell Technologies) containing 200 cells was plated in a well of a SmartDish (StemCell Technologies, 27370) in duplicates. The cells were incubated at 37C, for 14days. The resulting progenitor colonies were counted and scored using STEMVision Analysis (StemCell Technologies) according to the manufacturers instructions. The mean of the total number of colonies in the NTC samples for each experiment was set as 1.
Plasmid (6.5g) encoding WT hCD45RO or its variants was mixed with 200l serum-free DMEM medium and 19.5l polyethylenimine (1mgml1; Chemie Brunschwig, POL23966-100). The transfection mix was added dropwise to 1106 DF-1 cells plated the day before in a six-well plate. Cells were analysed 48h later by flow cytometry.
For in vitro ADC killing assays, 5,000 base-edited human activated T cells or 25,000 base-edited hCD34+ HSPCs were plated five days after electroporation in 96-well plates (flat-bottomed for T cells and round-bottomed for HSPCs; Corning 3596 and 3799, respectively) in 100l of corresponding medium (supplemented with only 50Uml1 of IL-2 for human T cells). For ADC killing assays involving saporin, a 100nM stock was prepared by incubating the biotinylated antibody (BC8 or MIRG451 mAbs) and saporinstreptavidin (ATS-Bio, IT-27-1000) at a 1:1 molar ratio for 30min at room temperature.
For in vitro ADC killing of co-cultures, 12,500 Jurkat cells were stained for 20min with CTV (Invitrogen, C34557A) at 37C and then seeded at a 1:1 cell ratio with 12,500 base-edited hCD34+ HSPCs five days post-electroporation in 96-well round-bottom plates in 100l HSPC medium with corresponding concentrations of CIM053SG3376 (ADC Therapeutics). Cells were incubated for 72h at 37C, stained for flow cytometry or cell sorting and analysed using a BD LSRFortessa. Genomic DNA was extracted for sequencing.
For in vitro ADC killing of mCherryluciferase-marked tumour cell lines (Jurkat, NALM-6, OCI-AML-2 and MOLM-14), 2,000 cells were plated in 384-well plates in medium with or without 30min pre-incubation at 37C with 50gml1 (333.33nM) naked CIM053 antibody (40l final total volume per well). Following a 72h incubation period, 5l firefly d-luciferin (0.75mgml1 resuspended in medium (Biosynth, L-8220)) was added to each well and incubated for 5min at room temperature. Luminescence readouts were recorded using a BioTek Synergy H1 plate reader.
For precise antibodyprotein affinity measurements and biophysical characterization, CD45wt and variants containing only D1 and D2 of the ECD were produced. The protein sequence (residues 225394) was histidine tagged at the carboxy terminus and contains few N- and C-terminal added amino acids (full WT sequence: ETGIEGRKPTCDEKYANITVDYLYNKETKLFTAKLNVNENVECGNNTCTNNEVHNLTECKNASVSISHNSCTAPDKTLILDVPPGVEKFQLHDCTQVEKADTTICLKWKNIETFTCDTQNITYRFQCGNMIFDNKEIKLENLEPEHEYKCDSEILYNNHKFTNASKIIKTDFGSPGEGTKHHHHHH, SEQ ID 57, Uniprot ID P08575). Expi293F GnTI cells (Thermo Fisher, A39240) that lack N-acetylglucosaminyltransferase I (GnTI) activity and therefore lack complex N-glycans were used for protein expression. After collection, the protein was purified using Ni-NTA chromatography followed by digestion of high-mannose glycans with endoglycosidase H (EndoHf; New England BioLabs, P0703S) at 37C overnight. EndoHf was removed from the protein solution with amylose resin and the CD45 protein was further purified by size-exclusion chromatography in buffer comprising 20mM HEPES, pH 7.4, 150mM NaCl. Peak monomer and dimer fractions (where needed) were concentrated using a 10kDa cut-off Amicon centrifugal filter (UFC8010) and protein aliquots were flash-frozen in liquid nitrogen before storage at 150C. Variant CD45 proteins were produced using the same experimental procedure. The monomer content percentage for each protein was taken from the size-exclusion chromatogram.
Analysis of MIRG451 and BC8 binding to the selected variants was performed on an Octet system RED96e (Sartorius) or R8 (Sartorius) at 25C with shaking at 1,000 rpm using 1 kinetic buffer (Sartorius, 18-1105). The selected variants were screened for their ability to bind to MIRG451 and BC8 using different concentrations of CD45 (WT or variant). MIRG451 was captured by an anti-human Fc-capture biosensor (AHC) (Sartorius, 18-5060) for 300s at 0.51gml1. As an analyte, human CD45wt and variants, containing only domains 1 and 2, were titrated at seven different concentrations, from 1,000nM to 15.6nM or from 50nM to 0.78nM with a 1:2 dilution series. Association of the analyte to MIRG451 was monitored for 600s and dissociation of the analyte from MIRG451 was monitored for 1,800s. Reference subtraction was performed against buffer-only wells. AHC tips were regenerated using 10mM Gly-HCl, pH 1.7. Data were analysed using the Octet data analysis software HT 12.0. Data were fitted to a 1:1 binding model. Kinetic rates Ka and Kd were globally fitted.
To analyse binding to BC8, streptavidin biosensors (Sartorius, 18-5020) were first coated with CaptureSelect biotin anti-LC- (murine) conjugate (Thermo Scientific, 7103152100) for 600s at 1gml1. BC8 was then captured by the coated streptavidin biosensors for 300s at 0.51.0gml1. Analyte titration was performed as for MIRG451. Association of the analyte to BC8 was monitored for 300s and dissociation of the analyte from BC8 was monitored for 900s. Reference subtraction, regeneration and data analysis were performed as for MIRG451.
The thermostability of CD45 D1D2 variants was analysed by differential scanning fluorimetry and monitoring tryptophane fluorescence using Nanotemper Prometheus NT.48 NanoDSF or a Nanotemper Prometheus Panta (NanoTemper Technologies)56,57,58. Thermal denaturation was monitored by tryptophane/tyrosine fluorescence at 350 and 330nm and an excitation wavelength of 280nm was used. CD45wt and variants were prepared at 0.251.0mgml1 in 20mM HEPES, 150mM NaCl, pH 7.4. Then 10l was put into the capillaries and placed into the sample holder. Each protein was measured in triplicates per experiment and the CD45wt was measured in four different experiments. The temperature was increased from 20C to 90C or 95C. The analysis was performed using the ratio of the fluorescent intensities at 350 and 330nm. The software of the instrument was used to calculate Tonset and TM as well as the mean and s.d. of the triplicates. The melting temperature was determined as the inflexion point of the sigmoidal curve and compared with that of CD45wt.
Flow cytometry was done on BD LSRFortessa instruments with BD FACSDiva software. Data were analysed with FlowJo software. Antibodies used for flow cytometry are listed in Supplementary Table 6. Cells were sorted with BD FACSAria or BD FACSMelody cell sorter instruments. Sorted cells were resuspended in 30l QuickExtract. PCRs were performed and sent for Sanger sequencing.
The CD45 expression of 27 people diagnosed with AML at University Hospital Basel was assessed using routinely acquired flowcytometry data as part of the diagnostic work-up. Gating for AML blasts, lymphocytes and erythrocytes was performed manually using FlowJo 10.10.0. Owing to the experimental set-up (threshold for SSC-A and FSC-A to exclude debris), a distinct erythrocyte population could not be distinguished in all samples (23 of 27). Data were analysed with GraphPad Prism 10 and statistical significance was calculated using mixed-effects analysis. All patients gave written informed consent to the analysis of clinical data for research purposes and the study was approved by the local ethics committee (BASEC-Nr 2023-01372).
All animal work was done in accordance with the federal and cantonal laws of Switzerland. Protocols were approved by the Animal Research Commission of the Canton of Basel-Stadt, Switzerland. All mice were housed in a specific pathogen-free condition in accordance with institutional guidelines and ethical regulations. NBSGW (stock 026622) female mice were purchased from Jackson Laboratories. HSPCs were edited as described above. Two days after electroporation, cells were collected and frozen in CryoStor CS10 medium. Cells were thawed on the day of injection, washed and resuspended in PBS. Recipient NBSGW female mice (4 weeks old) were injected intravenously into the tail vein with HSPCs (the number of cells injected varied between 0.6 and 1.1 million and is indicated in each figure). Chimerism was analysed by flow cytometry in blood after ten weeks. Mice were treated with saline or CIM053SG3376 at the dose(s) and intervals indicated in each figure. For tumour experiments in humanized mice, 1106 MOLM-14mCherryluc cells were injected into the tail vein. Then, 10 or 12 days after tumour inoculation, the mice were treated with saline or 1mg per kg CIM053SG3376. The mice received a second antibody dose of 0.5mg per kg CIM053SG3376 10 or 25 days after the first dose. Mice were euthanized 43 or 45 days after tumour inoculation or when reaching the maximum allowed clinical score. To monitor tumour growth, mice were injected intraperitoneally with 100l d-luciferin (BioSynth, L-8220) and were subjected to Newton7.0 imaging (Vilber).
For secondary transplant, NSGSGM3 female mice (stock 013062) were purchased from Jackson Laboratories. Recipient mice were irradiated the day before the BM transplant with 200cGy. Primary transplant mice were euthanized, the BM was isolated and 40% of it was re-injected into the new host. Mice from secondary transplants were euthanized 8 weeks after humanization.
MOLM-14mCherryluc (1106), OCI-AML-2mCherryluc (2106), JurkatmCherryluc (5106) or NALM-6mCherryluc (0.5106) cells were injected into the tail vein of NBSGW mice. After tumour inoculation, mice were monitored regularly (for behaviour, weight and imaging). Mice were treated with saline, control-SG3376 or CIM053SG3376 at the dose and intervals indicated in the relevant figure and euthanized 21 days after tumour inoculation or when reaching the maximum allowed clinical score.
Deidentified patient-derived AML samples were obtained from the PDX repository59,60 (Cancer Research Center of Toulouse, France). Signed written informed consent for research use in accordance with the Declaration of Helsinki was obtained from patients and approved by the Geneva Health Department Ethic Committee. PDX cells (0.6106) were injected into the tail vein of humanized NBSGW mice (8 weeks after HSPC injection). The weight of the mice was monitored regularly. Mice were treated with saline or CIM053SG3376 at the doses and intervals indicated in the figure and the mice were euthanized 54 days after tumour inoculation. Some control mice were euthanized 3 days before antibody treatment.
After the mice were euthanized, 0.2ml blood, both hind legs (femur and tibia) and the spleen were collected from each mouse. Cell suspensions were generated, red blood cells were lysed using ACK lysis buffer and then the cell suspensions were filtered. For tumour experiments, organs were collected on the day of euthanasia, single cell suspensions were generated and frozen in cryo medium. Samples from all mice were thawed and stained on the same day to minimize experimental variability. Cells were stained for different antigens and 30l Accucheck counting beads (1,066 microspheres per l; Invitrogen, PCB100) were added to each sample and the results were analysed by FACS using a BD LSRFortessa instrument.
Statistical analyses were done using GraphPad Prism 9 and 10 software. In all figure legends, n refers to the number of experimental replicates. For multiple comparisons, two-way ANOVA tests were used with significance levels indicated. Data are presented as meanstandard deviation. Survival curves were analysed using the log-rank MantelCox test. rhAmpSeq was analysed using a 2 test. The FDR was calculated using the BenjaminiHochberg method.
Some data points of the in vivo experiments were excluded after visual inspection of samples if the FACS time gate showed irregularities. One mouse that did not engraft HSPCs was excluded from Fig. 5 and Extended Data Fig. 10. Cell numbers in the sgNTC group treated with CIM053SG3376 were so low that analysis of some assays became unreliable (NGS, genetic chimerism analysis). We therefore excluded this group from NGS.
The number of biological replicates is specified for each experiment in the relevant figure legend. Several key experiments were performed by different people at times in different laboratories, and reagents were shared. For instance, identification of variants, characterization of recombinant variants and FACS validation were performed by different people. Some experiments were performed in the academic lab and validated in Cimeio labs and vice versa. To avoid unconscious bias when assigning mice to saline or the CIM053SG3376 groups, we always assigned the mice with the largest tumour mass to the ADC group.
The investigator who determined genetic chimerism (NGS and analysis) was blinded and provided the results to the investigator in charge of supervising in vivo experiments. The people who performed CHANGE-Seq_BE, rhAMPSeq and analysed the data were blinded and provided the results to the investigator in charge of supervising the in vivo experiments.
Non-proprietary materials are freely available on reasonable request. Restrictions apply to proprietary, commercial material.
Further information on research design is available in theNature Portfolio Reporting Summary linked to this article.
See the rest here:
Selective haematological cancer eradication with preserved haematopoiesis - Nature.com
Are any specific respiratory viruses more severe than others in recipients of allogeneic stem cell transplantation? A … – Nature.com
Piana JL, Prez A, Choro P, Guerreiro M, Garca-Cadenas I, Solano C, et al. Infectious Complications Subcommittee of the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH-TC). Respiratory virus infections after allogeneic stem cell transplantation: current understanding, knowledge gaps, and recent advances. Transpl Infect Dis. 2023;25:e14117 https://doi.org/10.1111/tid.14117.
Article CAS PubMed Google Scholar
Rachow T, Lamik T, Kalkreuth J, Kurze S, Wagner K, Stier P, et al. Detection of community acquired respiratory viruses in allogeneic stem-cell transplant recipients and controls-a prospective cohort study. Transpl Infect Dis. 2020;22:e13415 https://doi.org/10.1111/tid.13415.
Article CAS PubMed PubMed Central Google Scholar
Fontana L, Strasfeld L. Respiratory virus infections of the stem cell transplant recipient and the hematologic malignancy pa-tient. Infect Dis Clin North Am. 2019;33:52344. https://doi.org/10.1016/j.idc.2019.02.004.
Article PubMed PubMed Central Google Scholar
Seo S, Martin M, Xie H, Kuypers JM, Campbell AP, Choi SM, et al. Human rhinovirus RNA detection in the lower respiratory tract of hematopoietic cell transplant recipients: association with mortality. Biol Blood Marrow Transpl. 2013;19:S167S168. https://doi.org/10.1016/j.bbmt.2012.11.139.
Article Google Scholar
Ghosh S, Champlin R, Couch R, Englund J, Raad I, Malik S, et al. Rhinovirus infections in myelosuppressed adult blood and marrow transplant recipients. Clin Infect Dis. 1999;29:52832.
Article CAS PubMed Google Scholar
Kraft CS, Jacob JT, Sears MH, Burd EM, Caliendo AM, Lyon GM. Severity of human rhinovirus infection in immunocompromised adults is similar to that of 2009 H1N1 influenza. J Clin Microbiol. 2012;50:10613.
Article PubMed PubMed Central Google Scholar
Ogimi C, Waghmare AA, Kuypers JM, Xie H, Yeung CC, Leisenring WM, et al. Clinical significance of human coronavirus in bronchoalveolar lavage samples from hematopoietic cell transplant recipients and patients with hematologic malignancies. Clin Infect Dis. 2017;64:15329. https://doi.org/10.1093/cid/cix160.
Article CAS PubMed Google Scholar
Renaud C, Xie H, Seo S, Kuypers J, Cent A, Corey L, et al. Mortality rates of human metapneumovirus and respiratory syncytial virus lower respiratory tract infections in hematopoietic cell transplantation recipients. Biol Blood Marrow Transpl. 2013;19:12206. https://doi.org/10.1016/j.bbmt.2013.05.005.
Article Google Scholar
Seo S, Waghmare A, Scott EM, Xie H, Kuypers JM, Hackman RC, et al. Human rhinovirus detection in the lower respiratory tract of hematopoietic cell transplant recipients: association with mortality. Haematologica. 2017;102:112030. https://doi.org/10.3324/haematol.2016.153767.
Article CAS PubMed PubMed Central Google Scholar
Piana JL, Prez A, Montoro J, Hernani R, Lorenzo I, Gimnez E, et al. The effect of timing on community acquired respiratory virus infection mortality during the first year after allogeneic hematopoietic stem cell transplantation: a prospective epidemiological survey. Bone Marrow Transpl. 2020;55:43140. https://doi.org/10.1038/s41409-019-0698-7.
Article CAS Google Scholar
Piana JL, Prez A, Montoro J, Gimnez E, Gmez MD, Lorenzo I, et al. Clinical effectiveness of influenza vaccination after allogeneic hematopoietic stem cell transplantation: a cross-sectional, prospective, observational study. Clin Infect Dis. 2019;68:1894903. https://doi.org/10.1093/cid/ciy792.
Article CAS PubMed Google Scholar
Piana JL, Gmez MD, Prez A, Madrid S, Balaguer-Rosell A, Gimnez E, et al. Community-acquired respiratory virus lower respiratory tract disease in allogeneic stem cell transplantation recipient: risk factors and mortality from pulmonary virus-bacterial mixed infections. Transpl Infect Dis. 2018;20:e12926.
Article PubMed PubMed Central Google Scholar
Ljungman P, Tridello G, Piana JL, Ciceri F, Sengeloev H, Kulagin A, et al. Improved outcomes over time and higher mortality in CMV seropositive allogeneic stem cell transplantation patients with COVID-19; An infectious disease working party study from the European Society for Blood and Marrow Transplantation registry. Front Immunol. 2023;14:1125824 https://doi.org/10.3389/fimmu.2023.1125824.
Article CAS PubMed PubMed Central Google Scholar
Piana JL, Hernndez-Boluda JC, Calabuig M, Ballester I, Marn M, Madrid S, et al. A risk-adapted approach to treating respiratory syncytial virus and human parainfluenza virus in allogeneic stem cell transplantation recipients with oral ribavirin therapy: a pilot study. Transpl Infect Dis. 2017;19. https://doi.org/10.1111/tid.12729.
Seo S, Xie H, Campbell AP, Kuypers JM, Leisenring WM, Englund JA, et al. Parainfluenza virus lower respiratory tract disease after hematopoietic cell transplant: viral detection in the lung predicts outcome. Clin Infect Dis. 2014;58:135768.
Article PubMed Google Scholar
Shah DP, Ghantoji SS, Ariza-Heredia EJ, Shah JN, El Taoum KK, Shah PK, et al. Immunodeficiency scoring index to predict poor outcomes in hematopoi-etic cell transplant recipients with RSV infections. Blood. 2014;123:32638.
Article CAS PubMed PubMed Central Google Scholar
Austin PC. A tutorial on multilevel survival analysis: methods, models and applications. Int Stat Rev. 2017;85:185203. https://doi.org/10.1111/insr.12214.
Article PubMed PubMed Central Google Scholar
Piana JL, Xhaard A, Tridello G, Passweg J, Kozijn A, Polverelli N, et al. Seasonal human coronavirus respiratory tract infection in recipients of allogeneic hematopoietic stem cell transplantation. J Infect Dis. 2021;223:156475. https://doi.org/10.1093/infdis/jiaa553.
Article CAS PubMed Google Scholar
Nichols WG, Corey L, Gooley T, Davis C, Boeckh M. Parainfluenza virus infections after hematopoietic stem cell transplanta-tion: risk factors, response to antiviral therapy, and effect on transplant outcome. Blood. 2001;98:5738.
Article CAS PubMed Google Scholar
Chemaly RF, Hanmod SS, Rathod DB, Ghantoji SS, Jiang Y, Doshi A, et al. The characteristics and outcomes of parainfluenza virus infections in 200 patients with leukemia or recipients of hematopoietic stem cell transplantation. Blood. 2012;119:273845.
Article CAS PubMed Google Scholar
Piana JL, Tridelo G, Xhaard A, Wendel L, Montoro J, Vazquez L, et al. Upper and/or lower respiratory tract infection caused by human metapneumovirus after allogeneic hematopoietic stem cell transplantation. J Infect Dis. 2023;jiad268. https://doi.org/10.1093/infdis/jiad268.
Ljungman P, de la Camara R, Mikulska M, Tridello G, Aguado B, Zahrani MA, et al. COVID-19 and stem cell transplantation; results from an EBMT and GETH multicenter prospective survey. Leukemia. 2021;35:288594. https://doi.org/10.1038/s41375-021-01302-5.
Article CAS PubMed PubMed Central Google Scholar
Piana JL, Lpez-Corral L, Martino R, Vazquez L, Prez A, Martin-Martin G, et al. Infectious Complications Subcommittee of the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH-TC). SARS-CoV-2 vaccine response and rate of breakthrough infection in patients with hematological disorders. J Hematol Oncol. 2022;15:54. https://doi.org/10.1186/s13045-022-01275-7.
Piana JL, Heras I, Aiello TF, Garca-Cadenas I, Vazquez L, Lopez-Jimenez J, et al. Infectious Complications Subcommittee of the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH-TC). Remdesivir or Nirmatrelvir/Ritonavir Therapy for Omicron SARS-CoV-2 Infection in Hematological Patients and Cell Therapy Recipients. Viruses. 2023;15:2066 https://doi.org/10.3390/v15102066.
Article CAS PubMed PubMed Central Google Scholar
Chartrand C, Tremblay N, Renaud C, Papenburg J. Diagnostic accuracy of rapid antigen detection tests for respiratory syncytial virus infection: systematic review and meta-analysis. J Clin Microbiol. 2015;53:373849.
Article CAS PubMed PubMed Central Google Scholar
Aiello TF, Salmanton-Garcia J, Marchesi F, Weinbergerova B, Glenthoj A, Van Praet J, et al. Dexamethasone Treatment for COVID-19 is Related with Increased Mortality in Haematological Malignancy Patients: Results from the EPICOVIDEHA Registry. Available at SSRN: https://ssrn.com/abstract=4473151 or https://doi.org/10.2139/ssrn.4473151.
Regan P, Elkhalifa S, Barratt P. The systemic immunosuppressive effects of peripheral corticosteroid injections: a narrative review of the evidence in the context of COVID-19. Musculoskelet Care. 2022;20:43141. https://doi.org/10.1002/msc.1603.
Article Google Scholar
Piana JL, Lpez-Corral L, Martino R, Montoro J, Vazquez L, Prez A, et al. Infectious Complications Subcommittee of the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group (GETH-TC). SARS-CoV-2-reactive antibody detection after SARS-CoV-2 vaccination in hematopoietic stem cell transplant recipients: prospective survey from the Spanish Hematopoietic Stem Cell Transplantation and Cell Therapy Group. Am J Hematol. 2022;97:3042. https://doi.org/10.1002/ajh.26385.
Article CAS PubMed Google Scholar
Redjoul R, Le Bouter A, Beckerich F, Fourati S, Maury S. Antibody response after second BNT162b2 dose in allogeneic HSCT recipients. Lancet. 2021;398:298299. https://doi.org/10.1016/S0140-6736(21)01594-4.
Article CAS PubMed PubMed Central Google Scholar
Vasileiou S, Turney AM, Kuvalekar M, Mukhi SS, Watanabe A, Lulla P, et al. Rapid generation of multivirus-specific T lymphocytes for the prevention and treatment of respiratory viral infections. Haematologica. 2020;105:23543. https://doi.org/10.3324/haematol.2018.206896.
Article CAS PubMed PubMed Central Google Scholar
Mesenchymal Stem Cells Market to Witness an Outstanding Growth by 2030 – openPR
Mesenchymal Stem Cells Market
Major Highlights of TOC: Chapter 1: Overview of the Global Mesenchymal Stem Cells Market in 2024 1.1 GREEN HYDROGEN Industry Analysis 1.2 Key Companies and Product Profiles 1.3 Mesenchymal Stem Cells Market Segments 1.4 Industry Value Chain Analysis 1.5 Market Dynamics- Trends, Drivers, and Opportunities 1.6 Pricing Analysis 1.7 Porter's Five Forces Analysis 1.8 SWOT Profile 1.9 Macro-Economic and Demographic Impact Analysis 1.10 Scenario Analysis
Chapter 2: Global GREEN HYDROGEN Demand Forecasts 2.1 Overview of the Segment 2.2 Global Historic Mesenchymal Stem Cells Market Size (2018-2023) by Types, Applications, and Other Segments 2.3 Global Forecast Mesenchymal Stem Cells Market Size (2024-2030) by Types, Applications, and Other Segments
Chapter 3: Segment-wise Mesenchymal Stem Cells Market Forecasts 3.1 Key Market Segments 3.2 Premium Insights- Largest Types, Applications and Segments 3.3 Premium Insights- Most Lucrative Types, Applications, and Segments
Chapter 4: Mesenchymal Stem Cells Market Outlook by Country 4.1 Mesenchymal Stem Cells Market by Regions 4.2 Mesenchymal Stem Cells Market Revenue Share by Region 4.3 North America (US, Canada, Mexico) 4.4 Europe (Germany, UK, France, Spain, Italy, Russia, Others) 4.5 Asia Pacific (China, Japan, India, South Korea, Australia, South East Asia, Others) 4.6 Latin America (Brazil, Argentina, Chile, Others) 4.7 Middle East and Africa (Saudi Arabia, UAE, Qatar, South Africa, Nigeria, Egypt, Others)
Player Analysis in Chapter Five 5.1 Players' Market Share Analysis (2023) 5.2 Regional Market Concentration Rates 5.3 Business Profiles, SWOT Analysis, Financial Details, Product Portfolio of Companies ..........continued For a comprehensive competitive analysis, Buy this report now and gain access to a detailed table of contents @ https://www.usdanalytics.com/payment/report-9048 Why should you purchase this report? USD Analytics offers essential historical and analytical data on the global Mesenchymal Stem Cells Market. The report thoroughly evaluates future market trends and potential changes in market behavior. It provides various strategic business methodologies to support informed business decisions. Gain a competitive advantage in the market with this detailed research report, which covers competitive landscape analysis, growth drivers, applications, market dynamics, and other In conclusion, the Mesenchymal Stem Cells Market report is a genuine source for accessing the research data which is projected to exponentially grow your business. The report provides vital information including economic scenarios, benefits, limits, trends, market growth rates, and figures. Further, SWOT analysis and PESTLE analysis are also incorporated in the report. Review the Executive Report: @ https://www.usdanalytics.com/industry-reports/mesenchymal-stem-cells-market Thanks for reading this article; You can also get individual chapter-wise sections or region-wise report versions like North America, Middle East, Africa, Europe, MENA, LATAM, and Southeast Asia.
Contact Us: Harry (Business Consultant) USD Analytics Market Phone: +1 213-510-3499 sales@usdanalytics.com
About Author: USD Analytics is a leading information and analytics provider for customers across industries worldwide. Our high-quality research publications are connected market. Intelligence databases and consulting services support end-to-end support our customer research needs.
This release was published on openPR.
Continued here:
Mesenchymal Stem Cells Market to Witness an Outstanding Growth by 2030 - openPR
The Real Reason Studying Menstrual Blood Is So Important – Inverse
Roughly 20 years ago, a biologist named Caroline Gargett went in search of some remarkable cells in tissue that had been removed during hysterectomy surgeries. The cells come from the endometrium, which lines the inside of the uterus. When Gargett cultured the cells in a petri dish, they looked like round clumps surrounded by a clear, pink medium. But examining them with a microscope, she saw what she was looking for two kinds of cells, one flat and roundish, the other elongated and tapered, with whisker-like protrusions.
Gargett strongly suspected that the cells were adult stem cells rare, self-renewing cells, some of which can give rise to many different types of tissues. She and other researchers had long hypothesized that the endometrium contained stem cells, given its remarkable capacity to regrow itself each month. The tissue, which provides a site for an embryo to implant during pregnancy and is shed during menstruation, undergoes roughly 400 rounds of shedding and regrowth before a woman reaches menopause. But although scientists had isolated adult stem cells from many other regenerating tissues including bone marrow, the heart, and muscle no one had identified adult stem cells in the endometrium, Gargett says.
Such cells are highly valued for their potential to repair damaged tissue and treat diseases such as cancer and heart failure. But they exist in low numbers throughout the body and can be tricky to obtain, requiring surgical biopsy or extracting bone marrow with a needle. The prospect of a previously untapped source of adult stem cells was thrilling on its own, says Gargett. And it also raised the exciting possibility of a new approach to long-neglected womens health conditions such as endometriosis.
Before she could claim that the cells were truly stem cells, Gargett and her team at Monash University in Australia had to put them through a series of rigorous tests. First, they measured the cells ability to proliferate and self-renew and found that some of them could divide into about 100 cells within a week. They also showed that the cells could indeed differentiate into endometrial tissue and identified certain telltale proteins that are present in other types of stem cells.
Gargett, who is now also with Australias Hudson Institute of Medical Research, and her colleagues went on to characterize several types of self-renewing cells in the endometrium. But only the whiskered cells, called endometrial stromal mesenchymal stem cells, were truly multipotent, with the ability to be coaxed into becoming fat cells, bone cells, or even the smooth muscle cells found in organs such as the heart.
Around the same time, two independent research teams made another surprising discovery: Some endometrial stromal mesenchymal stem cells could be found in menstrual blood. Gargett was surprised that the body would so readily shed its precious stem cells. Since they are so important for the survival and function of organs, she didnt think the body would waste them by shedding them. But she immediately recognized the findings significance: Rather than relying on an invasive surgical biopsy to obtain the elusive stem cells shed identified in the endometrium, she could collect them via menstrual cup.
More detailed studies of the endometrium have since helped to explain how a subset of these precious endometrial stem cells dubbed menstrual stem cells end up in menstrual blood. The endometrium has a deeper basal layer that remains intact and an upper functional layer that sloughs off during menstruation. During a single menstrual cycle, the endometrium thickens as it prepares to nourish a fertilized egg, then shrinks as the upper layer sloughs away.
Gargetts team has shown that these special stem cells are present in both the lower and upper layers of the endometrium. The cells are typically wrapped around blood vessels in a crescent shape, where they are thought to help stimulate vessel formation and play a vital role in repairing and regenerating the upper layer of tissue that gets shed each month during menstruation. This layer is crucial to pregnancy, providing support and nourishment for a developing embryo. The layer and the endometrial stem cells that produce its growth also appear to play an important role in infertility: An embryo cant implant if the layer doesnt thicken enough.
Endometrial stem cells have also been linked to endometriosis, a painful condition that affects roughly 190 million women and girls worldwide. Although much about the condition isnt fully understood, researchers hypothesize that one contributor is the backflow of menstrual blood into a womans fallopian tubes, the ducts that carry the egg from the ovaries into the uterus. This backward flow takes the blood into the pelvic cavity, a funnel-shaped space between the bones of the pelvis. Endometrial stem cells that get deposited in these areas may cause endometrial-like tissue to grow outside of the uterus, leading to lesions that can cause excruciating pain, scarring, and, in many cases, infertility.
Researchers are still developing a reliable, noninvasive test to diagnose endometriosis, and patients wait an average of nearly seven years before receiving a diagnosis. However, studies have shown that stem cells collected from the menstrual blood of women with endometriosis have different shapes and patterns of gene expression than cells from healthy women. Several labs are working on ways to use these differences in menstrual stem cells to identify women at higher risk of the condition, which could lead to faster diagnosis and treatment. Menstrual stem cells may also have therapeutic applications. Some researchers working on mice, for example, have found that injecting menstrual stem cells into the rodents blood can repair the damaged endometrium and improve fertility.
Other research in lab animals suggests that menstrual stem cells could have therapeutic potential beyond gynecological diseases. In a couple of studies, for example, injecting menstrual stem cells into diabetic mice stimulated the regeneration of insulin-producing cells and improved blood sugar levels. In another, treating injuries with stem cells or their secretions helped heal wounds in mice.
A handful of small but promising clinical trials have found that menstrual stem cells can be transplanted into humans without adverse side effects. Gargetts team is also attempting to develop human therapies. She and her colleagues are using endometrial stem cells those taken directly from endometrial tissue rather than menstrual blood to engineer a mesh to treat pelvic organ prolapse, a common, painful condition in which the bladder, rectum, or uterus slips into the vagina due to weak or injured muscles.
The condition is often caused by childbirth. Existing treatments use synthetic meshes to reinforce and support weak pelvic tissues. However, adverse immune reactions to these materials have led these meshes to be withdrawn from the market. Gargetts research so far conducted only in animal models suggests that using a patients own endometrial stem cells to coat biodegradable meshes could yield better results.
Despite the relative convenience of collecting adult multipotent stem cells from menstrual blood, research exploring and utilizing the stem cells power and their potential role in disease still represents a tiny fraction of stem cell research, says Daniela Tonelli Manica, an anthropologist at Brazils State University of Campinas. As of 2020, she found, menstrual stem cell research accounted for only 0.25 percent of all mesenchymal cell research, while bone marrow stem cells represented 47.7 percent.
Manica attributes the slow adoption of menstrual stem cells in part to misogynistic ideas that uteruses are outside the norm and to reactions of disgust. Theres certainly something of an ick factor associated with menstrual blood, agrees Victoria Male, a reproductive immunologist at Imperial College London who coauthored an article about uterine immune cells in the 2023 Annual Review of Immunology.
Cultural taboos surrounding menstruation and a general lack of investment in womens health research can make it difficult to get funding, says Gargett. Immunologist Male has faced similar challenges it was easier to obtain funding when she used to study immune cells in liver transplantation than it is now that she works on immune cells in the uterus, she says.
If we want more research on menstrual fluid, we need more funding, says Male, noting that the logistics of collecting menstrual fluid over multiple days can be expensive. For that to happen, we have to tackle sex and gender bias in research funding. Through more equitable investments, she and others hope, menstruation will be recognized as an exciting new frontier in regenerative medicine not just a monthly inconvenience.
This article originally appeared in Knowable Magazine, an independent journalistic endeavor from Annual Reviews. Sign up for the newsletter.
Read the rest here:
The Real Reason Studying Menstrual Blood Is So Important - Inverse
Ongoing Research Aims to Expand Treatment Options, Address Unmet Needs in GVHD – OncLive
In an interview with OncLive, Mohamad Mohty, MD, PhD, discussed advancements in the management of graft-vs-host disease (GVHD) and highlighted unmet needs for patients with chronic (cGVHD) or acute GVHD (aGVHD), for whom standard-of-care therapies often fall short.
Mohty also expanded on ongoing research in GVHD reported at the 50th Annual EBMT Meeting. This included 3-year follow-up findings from the phase 2 ROCKstar trial (NCT03640481), which investigated belumosudil (Rezurock) in patients with cGVHD. The study showed that treatment with this agent yielded sustained responses, and no new safety signals were observed in eligible patients.1
Although challenges remain, advancements showcased at the EBMT meeting continue to underscore a new era for the treatment of patients with GVHD, Mohty noted. He provided further insights and key takeaways from the EBMT Annual Meeting in another interview with OncLive.
Mohty is a professor of hematology and the head of the Hematology and Cellular Therapy Department at Saint-Antoine Hospital and Sorbonne University in Paris, France. He is also head of the translational research team at the Saint-Antoine Research Center and the chairman of the Acute Leukemia Working Party of the EBMT.
Mohty: GVHD remains a matter of concern after transplant. Weve seen some major improvements over the past few years with the advent of posttransplant high-dose cyclophosphamide in particular. However, some patients are still [experiencing] aGVHD. Unfortunately, our first-line therapy remains focused on high-dose corticosteroids with deleterious adverse effects [AEs]; we also know that approximately 50% to 60% of patients are not going to respond to corticosteroids or are going to become corticosteroid dependent, which is not a good scenario.
Weve been lucky over the past 3 to 4 years to have the approval of the JAK2 inhibitor ruxolitinib, which proved to be a very good drug for patients who have steroid-refractory aGVHD. We are also struggling with a smaller group of patients who are becoming ruxolitinib refractory, resistant, or dependent. If you use ruxolitinib for salvage [therapy] after corticosteroids, a small group of patients may not be able to receive ruxolitinib, not everybody will respond, and some patients will lose their initial response to ruxolitinib. We then end up with a new group of patients who are ruxolitinib refractory. This is a new category of patients. Were fortunate that were now seeing research in this subgroup of patients. During the EBMT meeting, I was very excited to see some novel data on modulation of the microbiota in these patients with advanced and refractory aGVHD.
An important piece of information regarding the use of MaaT013 as salvage treatment based on the early-access program is its excellent safety profile. We didnt see severe AEs in the 140 patients who were treated. It is a single-shot treatment, which is important, instead of a treatment that is received and given to the patient on a chronic basis every day for several weeks or months.
Most importantly, the responses are very quickwithin 1 week. Patients who are responding are [doing so] in an excellent manner and very rapidly. Now based on this early-access program, we have very important follow-up that gives us some confidence in these results, which I consider quite reliable.
cGVHD incidence and severity have decreased over the past 10 years thanks to the efficacy of PTCy. However, we should bear in mind that were [performing transplant in] older patients. We are frequently using peripheral blood stem cells as well as donor lymphocyte infusion, including in the prophylaxis setting. At the end of the day, we still have a significant proportion of patients, at approximately 20%, who are continuing to experience moderate or severe cGVHD, which would require systemic immunosuppressive therapy.
Although cGVHD is not an immediate life-threatening condition, it dramatically alters the patients quality of life. Therefore, we still need to make progress in this arena. When it comes to the first-line therapy today, we dont have options better than corticosteroids. Thats the bad news because we dont like to use high-dose corticosteroids due to their deleterious AEs. We do have some good news about ruxolitinib because it was validated in the steroid-refractory cGVHD setting in the phase 3 REACH3 trial [NCT03112603].
But we have patients who may not be able to tolerate ruxolitinib; they may develop cytopenias over the long term or infections, or they may not respond to ruxolitinib. Therefore, novel options are needed, and this is where belumosudil, which was [granted regulatory] approved in 2021, is proving to be an excellent option. This drug has a new mechanism of action, as its a ROCK2 inhibitor. This mechanism of action is extremely interesting because when we look to the pathophysiology of cGVHD, we do have different phases. For instance, it is likely that ruxolitinib is acting well on the inflammatory component, which is an earlier phase, especially with the Th17 inflammatory response. But the use of a ROCK2 inhibitor such as belumosudil can also act not only on the inflammatory process but mainly on the fibrotic process. This is what makes it very attractive. This is what we saw in the ROCKstar trial, which allowed the approval of belumosudil in the United States and many other countries across the globe.
We can see more than a 70% response rate with belumosudil, and responses are in all organs usually involved with cGVHD, including the lung. [I stress] lung cGVHD because this is a terrible localization and [represents] a huge unmet need. Having a drug that [leads to] responses in the lung for cGVHD is most welcome. We are now getting closer to having better control of cGVHD. These drugs have been developed for the patient population with the most advanced, severe [disease]. The next step, and my wish, is to try to move these agents into earlier lines of use, even first-line treatment of cGVHD.
During the EBMT meeting we heard about the launch of a randomized trial with belumosudil in the first-line setting in combination with corticosteroids. The field is moving positively and, in my opinion, in the right direction.
Biomarkers in the field of GVHD, whether acute or chronic, are highly desired. I dont think anyone would question the need to have biomarkers to be able to predict the responses [and] also the failures, and maybe identify when to start treatment. Unfortunately, despite a lot of research and some very nice studies, Im not aware of 100% reliable, robust biomarkers in acute or chronic GVHD.
There have been some great work and results, for instance, with the Mount Sinai Acute GVHD International Consortium criteria and biomarker [research], but I still dont believe this is part of routine clinical practice. Its also true that in cGVHD, we dont have reliable biomarkers; maybe we will, but it will be difficult to find these biomarkers because cGVHD is a very heterogeneous condition. Clinically, cGVHD of the lungs or scleroderma may be different from dry eyes or dry mouth with cGVHD. In the ideal world, we would love to see these biomarkers. However, in the real world today, I dont see them ready for primetime yet.
Cutler C, Lee SL, Pavletic S, et al. Belumosudil for chronic graft-versus-host disease after 2 prior lines of systemic therapy: 3-year follow-up of the ROCKstar study. Presented at: 50th Annual Meeting of the EBMT; April 14-17, 2024; Glasgow, Scotland. Abstract OS13-01.
Go here to read the rest:
Ongoing Research Aims to Expand Treatment Options, Address Unmet Needs in GVHD - OncLive
Mohty Shares Key Takeaways From the 50th Annual EBMT Meeting, Highlighting Advances in GVHD – OncLive
At the 50th Annual EBMT Meeting, clinicians and researchers highlighted advancements in the blood and bone marrow transplant field, including significant advances centering on criteria for donor selection in allogeneic stem cell transplantation (ASCT) and ongoing efforts to refine outcomes and reduce complications in patients with graft-versus-host disease (GVHD), according to Mohamad Mohty, MD, PhD.
In an interview with OncLive, Mohty highlighted key takeaways from the meeting, emphasizing the discussions that revolved around donor selection criteria, as well as the impact that haploidentical donors and other donor sources may have on patients' needs.
The 2024 EBMT Meeting was a very exciting meeting; we celebrated the 50th anniversary of the European Society for Blood and Marrow Transplantation [EBMT], which was originally founded in 1974, Mohty said. This EBMT Annual Meeting proved to be very successful, and we [saw] the greatest advances in this field.
In the interview, Mohty discussed the implementation of post-transplant cyclophosphamide for GVHD prophylaxis, highlighted efforts to optimize cyclophosphamide dosing to minimize complications when treating this disease, and noted future directions for this research.
Mohty is a professor of hematology, as well as the head of the Hematology and Cellular Therapy Department at the Saint-Antoine Hospital and Sorbonne University in Paris, France. He is also a member and lead of the translational research team at the Saint-Antoine Research Center and is the chairman of the Acute Leukemia Working Party for the EBMT.
Mohty: What was striking for me were the criteria for choosing donors [and] the implementation and [expansion] of the use of posttransplant cyclophosphamide [PTCy] for GVHD prophylaxis. There are also novelties in the management of acute, especially steroid-refractory, but also ruxolitinib-refractory, GVHD. Weve seen some great advances when it comes to cGVHD, and overall, this was a memorable EBMT meeting where milestone data were presented.
When it comes to choosing a suitable stem cell donor [for] ASCT, there is a lot of excitement about the use of haploidentical donors. When combining matched sibling donors, fully matched unrelated donors, mismatched unrelated donors, and now haploidentical donors, youre able to find a suitable donor for approximately 95% of patients who need an ASCT. Although the use of cord blood cells has decreased over the past few years in the adult population, if you use cord blood cells [with] the new technologies and platforms for cord blood expansion, you would be able to cover approximately 100% of [patient] needs.
Interestingly, when it comes to living donors, the age of the donor remains an important parameter. Favoring a young donor, whether in the setting of haploidentical or unrelated donors, is likely to be an important parameter. Also, having a female donor to a male recipient remains an important risk factor for alloreactivity, especially in cGVHD. What remains controversial is how to choose between a young haploidentical donor, if available, and a young, fully matched, unrelated donor. Here we dont have a final response to this question.
There are teams who now rely almost exclusively on family donors, and this would include matched sibling donors and haploidentical donors. There are still teams who prefer to have fully matched 10 out of 10 unrelated donors. This is proving to be very exciting, and despite the debates, the good news is [there is a possibility of] finding a donor for almost everybody who needs an ASCT.
Age of the donor is important when selecting a given donoron the other hand, you must take into account the age of the recipient [too]. When it comes to the recipients age, it is very important to consider the comorbidities, which will play an important role in the final outcome. We know from daily clinical experience that a patient who is 60 years oldbut has comorbidities such as hypertension, diabetes, a heavy smoking history, and cardiovascular complications[will be affected by] these comorbidities. [They will] negatively impact the incidence of nonrelapse mortality.
However, a patient whos 70 years old but also fit and without any comorbidities [will] have a lower likelihood of developing toxicities. We can see an important separation between biological age vs physiological age, which is correlated with comorbidities. The development of comorbidity indexes and trying to use new electronic artificial intelligence tools to try to predict outcomes are going to be major [focuses] of future research.
The use of posttransplant high-dose cyclophosphamide has, in the past 15 years, proved to be a major breakthrough in ASCT because it has allowed safe haploidentical stem cell transplantation [to be performed for the first time, thus] allowing for engraftment while reducing the risk of severe aGVHD and cGVHD.
The use of PTCy has rapidly spread because it is a relatively cheap drug and physicians are quite familiar with the use of cyclophosphamide, as it is a very common drug in hematology and oncology. It has great efficacy in depleting the alloreactive T cells. Now there is an accumulating body of evidence explaining the mechanism of action that allows PTCy to effectively prevent GVHD.
[Although] the initial experience started in the haploidentical setting, use of PTCy is spreading beyond haploidentical transplant and is being used increasingly in the mismatched unrelated setting. Some teams are also using it in the unrelated matched sibling setting. These are the positives with use. We also know with some long-term follow-up and greater experience that high-dose PTCymay not be suitable or safe for everybody because there is a risk of cardiac complications, which were described especially in patients with cardiovascular risk factors. We also know [there is] the high risk of hemorrhagic cystitis related to the BK virus.
At the EBMT meeting, we saw very nice translational work showing that PTCy can impact the immune repertoire and immune anti-infectious controlthat can explain the higher incidence of infectious and opportunistic infections after PTCy. With this background, efforts are aiming to test lower doses of PTCy, and our team presented data on reducing the dose by 20% or even 30%.
For instance, [we used] 35 mg/kg per day for 2 days on day 3 and day 4 or day 3 and day 5. You can achieve similar GVHD prevention while reducing the risk of complications, especially cardiac [and] infectious complications, but also shortening the duration of neutropenia and thrombocytopenia. This needs to be validated in prospective future randomized trials, but the use of PTCywill continue to grow. This is something that will make the ASCT procedure safer and more feasible, especially in older and frail patients.
Mesenchymal stem cells pretreated with interferon-gamma attenuate renal fibrosis by enhancing regulatory T cell … – Nature.com
Cell culture
Human MSCs from bone marrow were purchased from Riken BioResource Research Center (Ibaraki, Japan)and cultured in Dulbeccos Modified Eagles Medium (DMEM: Sigma-Aldrich, St. Louis, MO, USA) with 10% fetal bovine serum (FBS, Sigma-Aldrich). Cells were passaged four to five times before use for transplantation. HK-2 cells, a human proximal tubular cell line, were obtained from the American Type Culture Collection (Manassas, VA). These cells were cultured as described previously47.
MSCs were pretreated with or without recombinant human IFN- (PeproTech, Cranbury, NJ, USA) by the following method. When MSCs reached 70% confluence, IFN- was added to the medium to achieve a final concentration of 10ng/mL. After 48h, cells were collected and subjected to in vivo and in vitro analyses.
Male Sprague Dawley (SD) rats (8weeks old) were purchased from Charles River Laboratories Japan (Yokohama, Japan). Experimental procedures were approved by the Institutional Animal Care and Use Committee of Hiroshima University (Hiroshima, Japan) (Permit Nos. A15-66 and A17-75) and conducted in accordance with the Guide for the Care and Use of Laboratory Animals, 8th ed, 2010 (National Institutes of Health, Bethesda, MD, USA). This study is reported in accordance with ARRIVE guidelines. To establish the animal model, SD rats were randomly divided in 6 groups (n=5 in each group): sham, PBS (control), MSCs, IFN- MSCs, NC siRNA/IFN- MSCs and IDO1 siRNA/IFN- MSCs groups. All procedures were performed under anesthesia with injection of agents composed of midazolam, medetomidine, and butorphanol. Right nephrectomy was performed 7days prior to IRI of the left kidney. Renal IRI was induced by transiently clamping the unilateral renal artery. After a laparotomy was performed, the left kidney was exposed. Next, the renal pedicle was clamped by atraumatic vascular clamps for 45min, followed by reperfusion on a heating blanket. After reperfusion, phosphate-buffered saline (PBS, vehicle), control MSCs, or IFN- MSCs (5105 cells/rat) were injected through the abdominal aorta clamped above and below the left renal artery bifurcation. At 7 or 21days post-injection, rats were sacrificed and their kidneys were collected to evaluate inflammation and fibrosis.
Immunohistochemical staining was performed according to previously described methods47 using the following primary antibodies: mouse monoclonal anti-Foxp3 (Abcam, Cambridge, UK), rabbit polyclonal anti-CD3 (Dako, Glostrup, Denmark), mouse monoclonal anti-rat CD68 (Serotec, Oxford, UK), and rabbit polyclonal anti-collagen type I (Abcam). FOXP3-, CD3- and CD68-positive cells, as well as areas positive for -SMA and collagen type I staining, were assessed using ImageJ software (version 1.53s, NIH) by examining five randomly selected fields (100magnification) of the cortex.
Double immunostaining was performed according to the following methods. Sections of formalin-fixed, paraffin-embedded tissues (4m thick) were de-paraffinized, subjected to heat-mediated antigen retrieval in citric acid buffer at 98C for 40min, and then blocked in 5% skim milk at room temperature for 1h. They were incubated with anti-FOXP3 antibody (Abcam) overnight at 4C, followed by incubation with the appropriate secondary antibody (DAKO) at room temperature for 1h, and then incubated with 3,3-diaminobenzidine (Sigma-Aldrich) at room temperature for 5min. After that, they were heated again in EDTA buffer (pH 9.0) in the same way. They were then blocked in 2.5% normal horse serum (ImmPRESS Horse Anti-Rabbit IgG Polymer kit; Vector Laboratories, Riverside, CA, USA) at room temperature for 20min, followed by incubation with anti-CD3 antibody (Abcam) overnight at 4C. They were incubated with the secondary antibody (ImmPRESS Horse Anti-Rabbit IgG Polymer kit; Vector Laboratories) at room temperature for 30min and then incubated with working solution prepared with Vector SG Peroxidase (HRP) Substrate Kit (Vector Laboratories) at room temperature for 5min.
Sections of formalin-fixed, paraffin-embedded tissues (2m thick) were stained with Massons trichrome to assess fibrosis. Areas of interstitial fibrosis were assessed using Lumina Vision (Mitani, Osaka, Japan) by examining five randomly selected fields (100magnification) of the cortex.
Sample collection and western blotting were performed as previously reported36,47 with the following primary antibodies: anti-VEGFA antibody (Abcam), mouse monoclonal anti--SMA (Sigma-Aldrich), rabbit monoclonal anti-TGF-1 (Abcam), IDO1 polyclonal antibody (Proteintech, Rosemont, IL, USA), mouse monoclonal anti-Foxp3 (Abcam), rabbit polyclonal anti-CD4 (Abcam), and mouse monoclonal anti-GAPDH (Sigma-Aldrich). Horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G (Dako) or goat anti-mouse immunoglobulin G (Dako) were used as secondary antibodies. SuperSignal West Dura or Pico Systems (Thermo Fisher Scientific, Waltham, MA, USA) were used to detect signals. The intensity of each band was analyzed by ImageJ software and standardized to the level of GAPDH.
To generate conditioned medium (CM) from untreated MSCs (control MSCs-CM) and IFN- MSCs (IFN- MSCs-CM), human MSCs (3105 cells/dish) were seeded in 10-cm dishes and cultured in DMEM containing 10% FBS. When the cells reached at least 70% confluence, the medium was replaced with fresh medium with or without 200ng/mL recombinant human IFN- (PeproTech). After 48h, the culture medium was replaced with DMEM containing 0.1% FBS, which was collected after 48h.
RNA extraction and real-time reverse-transcription PCR were conducted according to previously described methods47. Specific primers and probes for human IDO1 (assay ID: Hs00984148_m1), and human -actin (assay ID: Hs99999903_m1) were obtained as TaqMan Gene Expression Assays (Applied Biosystems, Foster City, CA, USA). mRNA levels were normalized to the level of -actin.
ELISA analysis of IDO (R&D Systems, Minneapolis, MN, USA) was performed according to the manufacturers protocol. Concentrations were normalized to the total protein content.
Human peripheral blood mononuclear cells (PBMCs; Biosciences, Berkeley, CA, USA) were suspended with the buffer formulated as MACS BSA Stock Solution (Miltenyi, Bergisch Gladbach, NRW, Germany) and autoMACS Rinsing Solution (Miltenyi). Cells were labelled with a Nave CD4+T Cell Isolation Kit II (Miltenyi) according to the manufacturers protocols. Nave CD4-positive T cells were sorted by negative selection using LS columns (Miltenyi) and MidiMACS (Miltenyi), and then collected.
Nave CD4 T cells (1106 cells/mL) were cultured in RPMI-1640 (Solarbio, Beijing, China) plus 0.1% FBS (Thermo Fisher Scientific) with MSCs-CM or IFN- MSCs-CM at a RPMI-1640:CM ratio of 1:1. Next, Dynabeads human T cell activator CD3/CD28 (Thermo Fisher Scientific) was added at a bead:cell ratio of 1:1, along with animal-free human recombinant IL-2 (ProteinTech) at a concentration of 300IU/mL, and cells were incubated in a humidified CO2 incubator. The medium, IL-2, and beads were exchanged on day 3, and then cells were collected on day 5.
MSCs were transfected with 20nM siRNA against IDO1 (s7426, Applied Biosystems) or negative control siRNA (4390843, Applied Biosystems) using Lipofectamine 2000 Transfection Reagent (Thermo Fisher Scientific). After 24h, transfected cells were washed and fresh complete medium was added. When cells reached 80% confluence, they were collected and subject to in vivo experiments.
Results are expressed as the meanstandard deviations (S.D.). For multiple group comparisons, one-way ANOVA followed by Bonferronis post-hoc test was applied. Comparisons between two groups were analyzed by Students t-test. P<0.05 was considered statistically significant.
All experimental procedures were approved by the Institutional Animal Care and Use Committee of Hiroshima University (Permit Nos. A15-66 and A17-75).
See original here:
Mesenchymal stem cells pretreated with interferon-gamma attenuate renal fibrosis by enhancing regulatory T cell ... - Nature.com
Stem Cell Treatment Restores Strength in Trial – Lifespan.io News
A placebo-controlled Phase 1/2 trial conducted in East Shanghai has found that administering umbilical cord-derived mesenchymal stem cells reduces frailty in older people.
These researchers begin by defining frailty as a state of heightened vulnerability to potential stressors as a consequence of reduction in physiological reserves across multiple systems [1]. This vulnerability destroys the strength and endurance of older people, exhausting their stamina and greatly increasing their risks of death and disability, and a metric has been determined to measure this [2]. However, while vitamin supplements may help people with nutritional deficiencies, there are no medically approved drugs to treat frailty [3].
As stem cell exhaustion has been pinpointed as a cause of frailty [4], replacement stem cells have been investigated as a possible treatment. In particular, mesenchymal stem cells (MSCs), which are naturally attracted to injury sites [5], appear to be the most promising. MSCs have multiple potential sources for derivation [6], and previous trials have been conducted to treat frailty by using MSCs derived from bone marrow (BM-MSCs), with positive results [7, 8].
This study, however, was conducted on stem cells that were originally derived from the human umbilical cord (HUC-MSCs). These cells are easy to mass produce [9], have been successfully clinically tested against other diseases such as heart failure [10] and arthritis [11], and fight inflammation [12]. This, however, is the first trial of HUC-MSCs for frailty.
All participants had to meet three criteria: to be between the ages of 60 and 80, to score between 1 and 4 on the Fried frailty scale [2], and to be expected to live another year. A large variety of co-morbidities were screened out, such as uncontrolled diabetes, serious cardiovascular problems, infections, and viral diseases. This was a double-blinded trial from which 80 potential candidates were excluded. 15 patients received placebo, and 15 received MSCs, for 6 months.
This study measured physical performance by testing grip strength, the timed up-and-go test, walking speed, and the ability to stand up and sit back down. Inflammatory cytokines such as interleukins were also measured, and sleep quality, quality of life, and mental health were also assessed.
There were no significant adverse effects. Three participants had suffered from ailments during the trial, two of which were in the placebo group and the third of which had dizziness not related to the MSCs.
Physical function, the primary endpoint of the study, was strongly affected by the MSCs. Even with only 30 total participants, not all of which participated in every assessment, the researchers were able to obtain, against baseline, a p-value of .003 after only one week of treatment and p-values under .001 for 1 and 6 months. Against placebo, the p-value at the end of the 6-month study was .042.
There were possible effects on mental health and sleep quality but those could be statistically attributed to the placebo effect. However, the treatment improved total quality of life with a p-value of 0.002 against placebo at the end of the study.
Cytokines had less clear effects; the placebo group spiked in TNF- and IL-17 at 6 months while the MSC group did not.
While not all of the endpoints were hit, this study was against frailty, and it is clear from these results that MSCs have beneficial impacts on frailty in human beings. However, this study was conducted in one country among 30 people. Further work, with a larger sample size and more testing sites, will need to be conducted to determine if these results hold up under further scrutiny.
To do this, we need your support. Your charitable contribution tranforms into rejuvenation research, news, shows, and more. Will you help?
[1] Clegg, A., Young, J., Iliffe, S., Rikkert, M. O., & Rockwood, K. (2013). Frailty in elderly people. The lancet, 381(9868), 752-762.
[2] Fried, L. P., Tangen, C. M., Walston, J., Newman, A. B., Hirsch, C., Gottdiener, J., & McBurnie, M. A. (2001). Frailty in older adults: evidence for a phenotype. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 56(3), M146-M157.
[3] Dent, E., Morley, J. E., Cruz-Jentoft, A. J., Woodhouse, L., Rodrguez-Maas, L., Fried, L. P., & Vellas, B. (2019). Physical frailty: ICFSR international clinical practice guidelines for identification and management. The Journal of nutrition, health and aging, 23(9), 771-787.
[4] Schulman, I. H., Balkan, W., & Hare, J. M. (2018). Mesenchymal stem cell therapy for aging frailty. Frontiers in Nutrition, 5, 108.
[5] Golpanian, S., Wolf, A., Hatzistergos, K. E., & Hare, J. M. (2016). Rebuilding the damaged heart: mesenchymal stem cells, cell-based therapy, and engineered heart tissue. Physiological reviews, 96(3), 1127-1168.
[6] Zhang, J., Huang, X., Wang, H., Liu, X., Zhang, T., Wang, Y., & Hu, D. (2015). The challenges and promises of allogeneic mesenchymal stem cells for use as a cell-based therapy. Stem cell research & therapy, 6, 1-7.
[7] Golpanian, S., DiFede, D. L., Khan, A., Schulman, I. H., Landin, A. M., Tompkins, B. A., & Hare, J. M. (2017). Allogeneic human mesenchymal stem cell infusions for aging frailty. Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences, 72(11), 1505-1512.
[8] Tompkins, B. A., DiFede, D. L., Khan, A., Landin, A. M., Schulman, I. H., Pujol, M. V., & Hare, J. M. (2017). Allogeneic mesenchymal stem cells ameliorate aging frailty: a phase II randomized, double-blind, placebo-controlled clinical trial. Journals of Gerontology Series A: Biomedical Sciences and Medical Sciences, 72(11), 1513-1522.
[9] Sarugaser, R., Lickorish, D., Baksh, D., Hosseini, M. M., & Davies, J. E. (2005). Human umbilical cord perivascular (HUCPV) cells: a source of mesenchymal progenitors. Stem cells, 23(2), 220-229.
[10] Bartolucci, J., Verdugo, F. J., Gonzlez, P. L., Larrea, R. E., Abarzua, E., Goset, C., & Khoury, M. (2017). Safety and efficacy of the intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure: a phase 1/2 randomized controlled trial (RIMECARD trial [randomized clinical trial of intravenous infusion umbilical cord mesenchymal stem cells on cardiopathy]). Circulation research, 121(10), 1192-1204.
[11] Wang, L., Huang, S., Li, S., Li, M., Shi, J., Bai, W., & Liu, Y. (2019). Efficacy and safety of umbilical cord mesenchymal stem cell therapy for rheumatoid arthritis patients: a prospective phase I/II study. Drug design, development and therapy, 4331-4340.
[12] Uccelli, A., Pistoia, V., & Moretta, L. (2007). Mesenchymal stem cells: a new strategy for immunosuppression?. Trends in immunology, 28(5), 219-226.
Read the original post:
Stem Cell Treatment Restores Strength in Trial - Lifespan.io News
Prayers For Baby Girl Born with Pale Purple Spots on Skin Winds Up Being Diagnosed with Two Different Types of … – SurvivorNet
27-year-old Kerri Paton was overjoyed to welcome her first child with husband Igor Topas, 28, a little over six years ago. Baby Amelia was born a healthy 6lbs 12oz. But shortly after Kerri gave birth, her doctor noticed the newborn had pale purple spots all over her body.
Once she was in my arms I just cried. I was so happy, Kerri recalls of the birth to UK news site yahoo!life.
Acute means that these types can progress rapidly.
RELATED: 11-Month-Old Baby Boy of Beloved Female Police Officer Is Diagnosed with AML Leukemia as Community Rallies in Support Fighting Pediatric Cancer
Amelia had to begin chemotherapy at just three weeks old. Luckily, after the first round of chemotherapy, Kerri saw that her babys lumps and spots were gone, though she still had a long way to go.
Amelia also endured a bone marrow transplant, or stem cell transplant, which is a procedure where healthy cells are transplanted into your blood or bone barrow.
After eight months of treatment, she was declared in remission by her medical team.
Six months later after hearing the joyous news of remission, during a routine follow-up, Kerri and Igor tragically learned that their baby girl had relapsed with AML. Igor had been on his way to the appointment when the doctor called Kerri and said she should come in as well.
I just knew it had come back, Kerry said.
What Are The Symptoms of Relapse in Acute Myeloid Leukemia (AML)?
In addition to more chemotherapy, Amelia needed another stem cell transplant.
Thankfully, the transplant worked, and Amelia has now been cancer free for five years. Just before Amelias recurrence, the Scottish couple had another baby, a son named Oscar, so theyre now a happy and healthy family of four.
Watching your kid sick it was horrible, Kerri said of all they have endured. At six years old, she noted that her daughter is just so funny and fiercely independent. Shes very strong-willed.
Added Kerri, Even though she went through cancer, her story doesnt end there.
Acute myeloid leukemia (AML) is a cancer that affects bone marrow, the spongy tissue inside of your bones. Its a rare cancer overall, but it is the most common type of leukemia in adults. Children rarely get AML.
Dr. Mikkael Sekeres, Director of the Cleveland Clinic Cancer Center Leukemia Program, Explains How AML Works
This disease is caused by DNA damage to the cells in your bone marrow that give rise to blood cells. Red blood cells carry oxygen to tissues, white blood cells fight infections as part of the immune system, and platelets help stop bleeding. Those cells are damaged in AML, and the damage results in an overproduction of unnecessary white blood cells.
Acute lymphoblastic leukemia (ALL) is a type of leukemia where the bone marrow makes too many immature lymphocytes, a type of white blood cell. It is also called acute lymphocytic leukemia, according to the National Cancer Institute.
Dr. Olalekan Oluwole, a hematologist with Vanderbilt University Medical Center, previously spoke with SurvivorNet about ALLs effect on the body and the type of treatments that work to fight it.
ALL is a type of cancer that is very aggressive, Dr. Oluwole told SurvivorNet. It grows very fast. Within a few weeks, a few months, the person will start to feel very sick. And thats why we will have to give it an equally aggressive type of treatment to break that cycle.
All About Acute Lymphoblastic Leukemia
Dr. Oluwole also says the leukemia often resides in the bone marrow, and because it is an abnormal growth, it just keeps dividing.
It doesnt follow rules, and it doesnt stop, he told SurvivorNet. Not only that, because this is part of the immune system, the immune system is sorta like the police of the body. So those abnormal cells that have now become cancer, they have the ability to go to many places. They go into the blood, and they often go into the tissue or the lining around the brain.
As parents navigate their young ones cancer journey, its important to remember that childrens bodies may react differently to treatments because their bodies are still growing.
They may receive more intense treatmentsand they may respond differently to drugs that control symptoms in adults, the National Cancer Institute informs. Be sure to ask a lot of questions.
RELATED: Why Do Pediatric Drugs Take So Long to Develop? A Look into the Lag Time on Drug Approvals for Childhood Cancer & Other Illnesses
Remember, youre not alone your childs oncologist and care team are there to guide you and provide information and answers. Oncological social workers can also be a vital resource to help you sort out the financial aspects of cancer treatment, as well as other cancer-related issues. Skilled psychologists and counselors can be accessed to help you maintain good mental health through your childs cancer journey, to the best of your ability.
Additionally, dont be afraid to reach out to your support system friends, relatives, etc. for help through this process. No one expects you to handle everything on your own.
Meanwhile, if youre wondering what you can do to ensure your child is getting the best treatment possible, consider the following recommendations from theNational Cancer Institute.
At SurvivorNet, we always encourage people to advocate for themselves when it comes to cancer and, more generally, healthcare. When it comes to a child, the parent must become the advocate.
RELATED: The Top Ten Childhood Cancer Symptoms That Can Be Missed
Its important to speak up about each and every issue that may concern you, no matter how minor, as even minor signs can sometimes clue doctors in on a potential cancer diagnosis. And catching it as early as possible is always ideal, as early detection may help with treatment and outcomes.
When It Comes to Health, Its Okay to Be a Little Pushy
Seeking multiple opinions is one way to make sure you or your child is getting the proper care and attention. You should also try to remember that not all doctors are in agreement. Recommendations for further testing or treatment options can vary, and sometimes its essential to talk with multiple medical professionals.
Every appointment you leave as a patient, there should be a plan for what the doc is going to do for you, and if that doesnt work, what the next plan is, Dr. Zuri Murell, director of the Cedars-Sinai Colorectal Cancer Center, previously told SurvivorNet. And I think that thats totally fair. And me as a health professional thats what I do for all of my patients.
Learn more about SurvivorNet's rigorous medical review process.
Mesenchymal Stem Cells Market Will Increase USD 10 Billion By 2033 – PharmiWeb.com
According to Market.us, the Mesenchymal Stem Cells Market Size is expected to achieve a value of around USD 10 billion by the year 2033. This indicates a noteworthy escalation from its 2023 valuation of USD 3 billion. Such substantial growth is forecasted to occur at a Compound Annual Growth Rate (CAGR) of 12.6% during the projection period spanning from 2024 to 2033.
The Mesenchymal Stem Cells Market is undergoing significant transformations, influenced heavily by its interconnectedness with various end-use industries. These industries are pivotal in shaping the Mesenchymal Stem Cells Markets dynamics, as they drive demand and set stringent quality standards. The alignment between the market offerings and the industries evolving needs ensures a consistent demand, fostering a scenario ripe for sustained growth in the Mesenchymal Stem Cells sector. This interdependence necessitates that market players remain agile, innovative, and responsive to the shifting requirements and emerging trends within these pivotal sectors.
Regulatory frameworks set by governments worldwide are integral to the Mesenchymal Stem Cells Markets structure, influencing its operational, environmental, and compliance standards. These regulations ensure the markets adherence to safety, quality, and sustainability norms, which are increasingly becoming stringent. The adherence to these standards in the Mesenchymal Stem Cells Market is not just about legal compliance but also about building trust with consumers and maintaining a competitive edge. The markets resilience is thus tied to its ability to navigate the complex regulatory landscape, adapt to new laws, and uphold the highest standards of operational excellence.
The Mesenchymal Stem Cells Markets dynamics are further shaped by the intricate import-export mechanisms and the flow of investments. Changes in trade policies, import-export regulations, and international tariffs directly influence the Mesenchymal Stem Cells Markets stability and growth trajectories. Investment from both governmental and private sectors plays a critical role, underpinning innovation and technological advancements in the Mesenchymal Stem Cells arena. These investments, along with strategic initiatives like mergers, acquisitions, and partnerships, are pivotal in driving the market forward, enabling scalability, and enhancing its global outreach.
Get sample: https://market.us/report/mesenchymal-stem-cells-market/request-sample/
In this market research, Market.us uncovered key insights that offer actionable takeaways and provide a clear direction for future market strategies. Mesenchymal Stem Cells market findings reveal critical trends and developments that shape the market landscape. These insights equip businesses with valuable information to make informed decisions and stay ahead of the competition. By understanding consumer preferences, market dynamics, and emerging opportunities, companies can optimize their product offerings, refine their marketing strategies, and capitalize on growth prospects. Mesenchymal Stem Cells research highlights the importance of staying agile and adaptable in response to evolving market conditions. With these key takeaways, businesses can confidently navigate the market landscape, mitigate risks, and drive sustainable growth in the long term.
Get quick access to our premium research report: https://market.us/purchase-report/?report_id=20920
In market research, its essential to identify and understand various market segments to tailor strategies effectively. By delineating the key market segments within the Mesenchymal Stem Cells market, businesses can refine their approach to cater to specific customer groups. This segmentation allows for more targeted marketing efforts, product development, and customer relationship management. Through thorough analysis, industries can identify common characteristics, needs, preferences, and behaviors within each segment. Mesenchymal Stem Cells insights enable companies to craft tailored messaging, promotions, and offerings that resonate with the unique needs of each segment. Moreover, understanding Mesenchymal Stem Cells market segments facilitates resource allocation, helping businesses allocate their resources efficiently and maximize their return on investment. Overall, identifying and targeting key market segments is crucial for businesses seeking to effectively engage with their target audience and achieve sustainable growth.
Request a sample copy of the report: https://market.us/report/mesenchymal-stem-cells-market/request-sample/
When researching the Mesenchymal Stem Cells industry, its crucial to understand and leverage factors that drive growth. These may include technological advancements, increasing consumer demand, and supportive government policies. By recognizing and capitalizing on these forces, Mesenchymal Stem Cells industry can position themselves strategically to capitalize on growth opportunities. However, its also important to address market restraints such as regulatory challenges, economic downturns, and shifting consumer preferences. By identifying these obstacles early on, businesses can develop strategies to mitigate their impact and navigate through challenges effectively.
Additionally, exploring untapped Mesenchymal Stem Cells market opportunities and emerging trends is essential. This involves identifying new market segments or niche markets and developing targeted strategies to capture these opportunities. Staying informed about Mesenchymal Stem Cells market trends, including shifts in consumer behavior, technological innovations, and the competitive landscape, is crucial for maintaining a competitive edge. Overall, comprehensive market research involves analyzing internal and external factors to make informed decisions and drive sustainable growth within the Mesenchymal Stem Cells industry.
The regional analysis of the Mesenchymal Stem Cells market provides valuable insights into its performance across various geographical areas, offering a comprehensive understanding of the opportunities and challenges present in each region. By examining factors such as economic conditions, regulatory frameworks, consumer preferences, and competitive landscapes, researchers can identify key trends and dynamics shaping Mesenchymal Stem Cells market dynamics at the regional level. This analysis enables stakeholders to tailor their strategies and investments to capitalize on specific market nuances and maximize growth potential. Moreover, understanding regional variations allows companies to mitigate risks associated with Mesenchymal Stem Cells market fluctuations and adapt their approaches to effectively target diverse customer segments. Overall, a robust regional analysis serves as a vital tool for informed decision-making and successful market penetration strategies.
To tailor this report to your needs, lets delve deeper into the specifics of the report: https://market.us/report/mesenchymal-stem-cells-market/#inquiry
In assessing the competitive landscape of the Mesenchymal Stem Cells market, it is essential to analyze key players strengths, weaknesses, and strategies. Leading companies in the healthcare sector typically have robust distribution networks, strong brand recognition, and diversified product portfolios, which are their primary strengths. However, they may also face challenges such as fluctuating market demand, regulatory constraints, and competitive pricing pressures. Strategies employed by Mesenchymal Stem Cells industry players often include product innovation, strategic partnerships, mergers and acquisitions, and market expansion initiatives. By continuously leveraging their strengths and addressing weaknesses, these companies strive to maintain or enhance their market position while adapting to evolving industry dynamics. A comprehensive understanding of the competitive landscape enables stakeholders to make informed decisions and develop effective strategies to capitalize on Mesenchymal Stem Cells market opportunities.
Recent developments in the Mesenchymal Stem Cells market, including mergers, acquisitions, and product launches, are shaping the industry landscape. These events reflect strategic maneuvers by companies to gain competitive advantage and expand their market presence. Mergers and acquisitions often lead to market consolidation and portfolio diversification, while new product launches drive innovation and address evolving consumer demands. Staying informed about Mesenchymal Stem Cells developments is crucial for understanding market dynamics and identifying opportunities for growth and investment.
Dont miss out on this valuable resource! Order your copy of the report today and unlock the full potential of this thriving industry!: https://market.us/purchase-report/?report_id=20920
Contact Details Market.us (Powered By Prudour Pvt. Ltd.) Contact No: +1 7186184351. Email: inquiry@market.us Blog: https://medicalmarketreport.com/
View More Trending Reports
Healthcare Cleanroom Consumables Market Projected to reach USD 17.1 Bn with 5.3% CAGR By 2033
Hernia Repair Devices Market Predicted USD 9 Billion by 2033, An Approximate 4.1% CAGR Growth
Joint Replacement Market Will Increase USD 26.5 Billion By 2033 and 4.3% CAGR
Lip Augmentation Market Projected To Reach USD 14.8 Billion With 7.9% CAGR By 2033
Orthopedic Navigation Systems Market Will Increase USD 8.9 Billion By 2033 with 12.7% CAGR
Cardiovascular and Soft Tissue Repair Patches Market
Ursodeoxycholic Acid Drug Product Market
Treatment Planning Systems And Advanced Image Processing Market
Custom Antibody Services Market
Lateral Flow Readers Market
Read the original:
Mesenchymal Stem Cells Market Will Increase USD 10 Billion By 2033 - PharmiWeb.com
VRD versus VCD as induction therapy before autologous stem cell transplantation in multiple myeloma: a nationwide … – Nature.com
Induction therapy followed by autologous stem cell transplantation (ASCT) is standard of care for young and fit patients with newly diagnosed multiple myeloma (MM) [1]. Induction therapy has evolved from doublet to triplet to quadruplet regimens over the last decades. The most common triplet therapy is either Bortezomib-Cyclophosphamide-Dexamethasone (VCD), Bortezomib-Lenalidomide-Dexamethasone (VRD), or less frequently Bortezomib-Thalidomide-Dexamethasone (VTD). No large, randomized phase III study comparing the VCD and VRD regimens has been conducted and is unlikely to be done in the future. Retrospective studies and smaller prospective studies comparing VRD and VCD have produced mixed results [2,3,4,5,6,7].
In Norway, there has been a shift from VCD to VRD induction therapy in recent years, while VTD has only been used in a minority of patients. ASCT for multiple myeloma in Norway is performed at four centers, and comprehensive population-based nationwide follow-up data are available from electronic journals.
In collaboration with all centers in Norway doing ASCT, we identified all patients in Norway who had undergone ASCT for multiple myeloma in the study period 2008 to 2020.
We included patients with multiple myeloma [8] who received first line induction therapy followed by ASCT in the period from January 1st 2008 to December 31st 2020 in Norway. We did not include patients who received induction therapy but did not proceed to ASCT. Patients were censored March 1st 2022 or at loss to follow-up because of relocation outside of Norway (n=5), or if the journal from the local hospital could not be obtained (n=7).
Data was collected from electronic patient journals at the transplant centers and from hospitals responsible for induction therapy and follow-up after ASCT. Change of induction therapy was recorded if a patient changed from one line to another, and the reason for change was collected. Patients who changed therapy were not included in the primary response analysis, regardless of the reason for change, but they were included in a separate intention-to-treat analysis. All patients, including those who changed treatment, were included in the PFS and OS analysis. Further description of study design, endpoints and statistical analysis is provided in the supplementary material.
We identified 1354 patients who received ASCT as first-line treatment for multiple myeloma in Norway in the study period.
Of these, 682 patients received VCD induction, 332 patients received VRD induction, and 42 patients received VTD induction. Baseline characteristics are described in Table 1 and were largely similar between patients who received VCD and VRD induction, with two notable exceptions. Patients in the VRD group were older than patients in the VCD group (median 62 years vs. 60 years). Patients in the VRD group received ASCT in more recent years (mostly 2017-2020), compared to VCD.
Three months after ASCT, response rates were higher with VRD, with 89% in the VRD group achieving VGPR, versus 76% in the VCD group (p<0.001). In the intention-to-treat analysis, the difference in response between the groups remained statistically significant (Table 1).
In the VCD group, 4% of patients changed therapy due to lack of response, and 1% due to progression. In the VRD group, very few patients changed treatment due to lack of response (1%) or progression (1%). Only a small minority, 3% and 2% of patients in the VRD and VCD group respectively, changed treatment due to adverse effects (Table 1). In patients who received bortezomib, thalidomide and dexamethasone (VTD), 36% of patients changed treatment due to side effects (Supplementary Table 5).
Patients in the VRD group more often received treatment after ASCT than in the VCD group (61% vs. 14%, p<0.001). Consolidation treatment (22% vs. 1%), maintenance treatment (25% vs. 10%) or both (14% vs. 3%) were all more frequent in the VRD group (Table 1). In the VCD group, 4.7% of patients (n=32) and in the VRD group 3.9% of patients (n=13) had progressive disease before starting consolidation or maintenance treatment.
The median follow-up time of patients still alive at data cut-off was 79 months (range: 19179 months) in the VCD group and 38 months (range: 1871 months) in the VRD group.
In the VCD group, the median PFS was 30.1 months (95% confidence interval (CI) 28.331.9 months). In the VRD group, the median PFS was 55.1 months (95% CI 46.0-not reached (NR), Fig. 1A). The difference was significant on log-rank test, p<0.001. In the VTD group median PFS was 36.6 months (Supplementary Fig. 2)
A PFS, all patients. B PFS, only patients who received maintenance treatment. C PFS, only patients who revied ASCT in later years (20172020) and did not received any post-ASCT treatment. D OS, all patients.
When we included only patients who received maintenance therapy after ASCT, the median PFS in the VCD group increased to 47.1 months, which is not statistically different from patients in the VRD group who received maintenance, who had a median PFS of 56.4 months, p=0.174 (Fig. 1B).
In a separate analysis excluding patients who received maintenance and/or consolidation and who received ASCT in later years (20172020), VRD was superior to VCD regarding PFS with a log-rank test of p<0.001 (Fig. 1C).
The median OS for VCD was 114.0 months (95% CI 103.4125.8 months) and the median OS for VRD was not reached, log-rank test p<0.001 (Fig. 1D).
The hazard ratios for PFS and OS on multivariate analysis is provided in Supplementary Table 2. After controlling for patient and disease factors, VCD had inferior PFS compared to VRD (HR 2.08, 95% CI 1.492.91, p<0.001). There was no significant difference in OS between the two regimens in multivariate analysis.
VTD is approved by the European Medical Agency as induction therapy before ASCT. This is not the case for VCD and VRD, although they are used widely in current clinical practice. The most recent European Society of Medical Oncology (ESMO) guidelines [1] recommend VRD as the first option for induction therapy. Daratumumab-VTD is also approved and recommended, but our study confirms the high toxicity associated with regimens containing thalidomide. VRD is the comparator arm in recent clinical trials comparing Daratumumab-VRD vs VRD before ASCT [9, 10]. Our study supports the use of VRD in both clinical practice, and as the standard treatment arm in clinical trials, as it is more effective than VCD and better tolerated than VTD. However, recent results from the PERSEUS trial [9], with significantly longer PFS for Daratumumab-VRD vs VRD, will most likely be practice changing.
We observed a statistically significant improvement in both PFS and OS favoring VRD. This must, however, be interpreted with caution. The difference in use of post-ASCT therapy, and the time periods in which the regimes were given, are two major biases. We corrected for this by performing a separate analysis for only patients who received ASCT in later years and who did not receive consolidation and/or maintenance therapy. In this analysis VRD still showed significantly longer PFS compared to VCD. The median PFS was also longer in the VRD group when only patients who received maintenance therapy were included, although the difference was not statistically significant. Multivariate analysis showed a statistically significant PFS benefit favoring VRD, but no statistically significant OS benefit. Given the median overall survival in our data of approximately 9.5 years in the VCD group, induction therapy administered for 24 months represents only a fraction of this total observed time. Therefore, the effect of induction therapy on overall survival may be modest, and other factors, like treatment options available at relapse, will have a significant impact on patient survival. Most patients in the VCD group received ASCT before 2017, when consolidation and maintenance treatment were uncommon and fewer treatment options were available at relapse, affecting the survival of this group negatively. Conversely, in the VRD group, most patients received ASCT after 2017. In this period, maintenance treatment was usually given (or consolidation treatment when maintenance was not reimbursed), and effective treatments like CD38-antibodies and carfilzomib were available at relapse.
The main limitation of the study is its retrospective nature, and as patients were not randomized, confounding factors cannot be excluded. However, the type of induction the patient received was mainly dependent on center and not on patient or disease factors. Standard induction therapy differed between regions, where some centers consistently used VCD while others consistently used VRD. In Norway, access to new therapies is similar for all, and national and regional treatment guidelines are usually the factors that determine choice of treatment, and to a lesser degree individual patient risk factors. Furthermore, a limitation was that we only included patients who received ASCT. Patients who died before ASCT, started induction therapy but for various reasons did not proceed to ASCT, including those who could not harvest enough stem cells, were not included. The follow-up time for VRD patients was relatively short. Patients were included from many different hospitals in Norway over a long period of time, with variable practices regarding timing of treatment start, dosing schedules, response assessment and supportive care. Although the data quality was generally good, some data was missing and incomplete.
Our study is the first to report from a comprehensive nationwide, population-based cohort with a very low proportion of patients lost to follow-up. This is a major strength, as the inclusion of a broad, heterogenous population increases the generalizability of the results and reduces the risk of selection bias. Our data included an overlap period where both regimens were given. Apart from the type of induction therapy, the treatment course between the two groups were similar; Length of induction treatment, the ASCT procedure and time to response evaluation was unchanged throughout the study period and similar for both groups.
In conclusion, our results suggests that VRD should be preferred to VCD as induction therapy for newly diagnosed MM patients who are eligible for ASCT.
Original post:
VRD versus VCD as induction therapy before autologous stem cell transplantation in multiple myeloma: a nationwide ... - Nature.com
Unlocking the healing potential of dental stem cells – Varsity
In recent years, the field of regenerative medicine has witnessed remarkable advancements in the use of dental stem cells. The discovery of these tiny but potent cells nestled within our teeth has sparked excitement among researchers and medical professionals alike, offering new avenues for treating a myriad of diseases and injuries.
A journey through stem cell history
Stem cells are cells that have the unique ability to develop into various specialized cell types in the body. This capacity for differentiation is what makes them invaluable in regenerative medicine.
The history of stem cell research traces back to the mid-twentieth century. In the 1960s, Canadian scientists James Till and Ernest McCulloch conducted experiments demonstrating the existence of stem cells in bone marrow, the spongy tissue inside bones that can produce blood cells, after introducing new cells into the bone marrow of irradiated mice. This work laid the foundation for further exploration into the therapeutic potential of stem cells.
Since then, researchers have identified stem cells in various tissues throughout the body, including in dental pulp, the soft tissue inside teeth. Dental stem cells have garnered particular interest due to their easy accessibility and regenerative capabilities.
Within the dental pulp, researchers have identified several types of stem cells, each with its own unique properties and potential applications.
Dental pulp stem cells
Dental pulp stem cells (DPSCs) are stem cells found in the centre of teeth. They have unique abilities to grow and transform into different types of cells, like those found in teeth, nerves, bones, muscles, and even certain organs. These cells can help in repairing damaged teeth and treating various diseases.
DPSCs are isolated from dental pulp tissue, typically obtained from third molars, also known as wisdom teeth, that are often extracted and discarded. The isolation process does not involve invasive surgical procedures and poses no harm to the donor.
DPSCs have the ability to repair damaged or diseased dental tissues. They can become tooth-building cells called odontoblasts cells that form dentin, the hard tissue beneath the enamel and osteoblasts cells that build jaw bone. This could revolutionize dental treatments by enabling the regeneration of natural tooth structures, reducing the need for fillings, crowns, and other restorative procedures.
DPSCs may even be able to help treat several medical conditions. In nerve-related conditions, such as nerve trauma or neurodegenerative diseases, patients may experience a loss or damage of neurons that need replacement or repair. DPSCs can help as they possess the ability to differentiate into neuron-like cells. Surprisingly, they can also secrete neurotrophic factors, which are biomolecules that support the growth, survival, and function of neurons and promote neuronal repair.
In conditions where the immune system is overactive such as inflammatory or autoimmune diseases, or neuroinflammation in neurodegenerative diseases such as Parkinsons disease DPSCs can help calm down the immune system by releasing molecules that reduce viral replication and reduce inflammation produced by the immune system.
Additionally, DPSCs have also shown promise in repairing damaged heart tissue and improving blood flow in conditions like heart attacks and leg artery blockages, including by stimulating formation and repair of blood vessels.
Stem cells from human exfoliated deciduous teeth
In 2003, researchers discovered a variety of stem cells in shed baby teeth, which are scientifically known as human exfoliated deciduous teeth. These cells have the remarkable ability to transform into various cell types like bone, nerve, and liver cells and can specialize into other types of stem cells.
When transplanted into living organisms, stem cells from human exfoliated deciduous teeth (SHEDs) show potential in repairing bone defects and forming new dental tissue.
SHEDs also possess immune-regulating properties according to studies in mice, and they could be beneficial for treating diseases like lupus by balancing the immune response. This means that we may be able to treat lupus, which is a genetic disorder characterized by the inflammation of different tissues, with stem cells used to repair damaged nerves and slow progression. This treatments success is proportional to a patients age and duration of condition.
Immature dental pulp stem cells
In recent studies, researchers have identified a special type of dental stem cells known as Immature Dental Pulp Stem Cells (IDPSC), also found in the dental pulp of baby teeth. These cells express certain markers that indicate they are at an early stage of development and are markers in embryonic stem cells.
One exciting discovery is that when researchers transplanted cell sheets made from undifferentiated IDPSC into rabbits with damaged corneas the outermost layer of the eye they observed the regeneration of the outer layer of the cornea. This finding suggests potential applications in treating corneal injuries and reconstruction.
Moreover, experiments involving the transplantation of IDPSC into immunocompromised mice and dogs have demonstrated promising results. The transplanted IDPSC were able to integrate well into various tissues and significantly improve conditions such as muscular dystrophy in dogs, without triggering immune rejection.
Harnessing the healing power of dental stem cells
The versatility of dental stem cells holds immense promise for regenerative medicine. Researchers are exploring a wide range of potential applications, including dental regeneration, bone regeneration, and neurological disorders.
As research continues to advance, we may soon see these tiny but mighty cells transform the landscape of healthcare, offering new hope for patients suffering from a wide range of conditions. From repairing damaged teeth to restoring neurological function, the possibilities seem to be truly endless.
Read the original post:
Unlocking the healing potential of dental stem cells - Varsity
Key mechanism controlling bone marrow stem cells could lead to new therapies – PharmaTimes – PharmaTimes
Hoxa9 and b-catenin molecules are a rare population of self-renewing HSCs found in bone marrow
Researchers from Kings College Londons (KCL) Comprehensive Cancer Centre have identified a key mechanism that governs how bone marrow stem cells work, which could potentially lead to new therapeutic pathways.
The findings from the study will help researchers further understand the key principles involved in stem cell biology and could provide new avenues for the development of efficient stem cell therapeutics.
Researchers identified two molecules, Hoxa9 and b-catenin, that control when bone marrow stem cells rest and recover, as well as when they act and replicate.
Both molecules work together to control a rare population of self-renewing stem cells that are predominantly found in bone marrow, known as haematopoietic stem cells (HSCs).
HSCs are protected from environmental stressors and prevent exhaustion by resting, while inactive HSCs must become active again, replicating themselves by turning into different blood cells, including red blood cells, white blood cells and platelets, to replenish the blood system and respond to problems including infections, blood loss and other complications.
Researchers discovered that this active/inactive characteristic of HSCs plays a key role in bone marrow transplantation, a vital procedure for several diseases, including cancer, as it is critical for cancer stem cells, which sustain the disease and cause relapse.
Researchers suggest that understanding this process will be vital when designing better treatments.
In addition, the team identified a critical enzyme known as PRMT1, which mediates the functions of the two molecules, offering a potential new avenue for the development of efficient stem cell therapeutics.
Eric So, professor and chair in Leukaemia Biology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, KCL and study lead, commented: Given the critical functions of stem cells in bone marrow transplant and cancer biology, [the] identification of a new druggable pathway not only will help to better understand the stem cell biology but also facilitates the development of more effective therapeutics in the future.
Addition of ruxolitinib to standard graft-versus-host disease prophylaxis for allogeneic stem cell transplantation in … – Nature.com
Georges GE, Doney K, Storb R. Severe aplastic anemia: allogeneic bone marrow transplantation as first-line treatment. Blood Adv. 2018;2:20208.
Article CAS PubMed PubMed Central Google Scholar
Zhao J, Ma L, Zheng M, Su L, Guo X. Meta-analysis of the results of haploidentical transplantation in the treatment of aplastic anemia. Ann Hematol. 2023;102:256587.
Article CAS PubMed Google Scholar
Esteves I, Bonfim C, Pasquini R, Funke V, Pereira NF, Rocha V, et al. Haploidentical BMT and post-transplant Cy for severe aplastic anemia: a multicenter retrospective study. Bone Marrow Transplant. 2015;50:6859.
Article CAS PubMed Google Scholar
Xu LP, Jin S, Wang SQ, Xia LH, Bai H, Gao SJ, et al. Upfront haploidentical transplant for acquired severe aplastic anemia: registry-based comparison with matched related transplant. J Hematol Oncol. 2017;10:25.
Article PubMed PubMed Central Google Scholar
Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799807.
Article CAS PubMed PubMed Central Google Scholar
Mart-Carvajal AJ, Anand V, Sol I. Janus kinase-1 and Janus kinase-2 inhibitors for treating myelofibrosis. Cochrane Database Syst Rev. 2015;2015:CD010298.
PubMed PubMed Central Google Scholar
Spoerl S, Mathew NR, Bscheider M, Schmitt-Graeff A, Chen S, Mueller T, et al. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood. 2014;123:383242.
Article CAS PubMed Google Scholar
Schroeder MA, Choi J, Staser K, DiPersio JF. The role of Janus kinase signaling in graft-versus-host disease and graft versus leukemia. Biol Blood Marrow Transplant. 2018;24:112534.
Article CAS PubMed Google Scholar
Hechinger AK, Smith BA, Flynn R, Hanke K, McDonald-Hyman C, Taylor PA, et al. Therapeutic activity of multiple common -chain cytokine inhibition in acute and chronic GVHD. Blood. 2015;125:57080.
Article CAS PubMed PubMed Central Google Scholar
Choi J, Ziga ED, Ritchey J, Collins L, Prior JL, Cooper ML, et al. IFNR signaling mediates alloreactive T-cell trafficking and GVHD. Blood. 2012;120:4093103.
Article CAS PubMed PubMed Central Google Scholar
Carniti C, Gimondi S, Vendramin A, Recordati C, Confalonieri D, Bermema A, et al. Pharmacologic inhibition of JAK1/JAK2 signaling reduces experimental murine acute GVHD while preserving GVT effects. Clin Cancer Res. 2015;21:37409.
Article CAS PubMed Google Scholar
Zeiser R, von Bubnoff N, Butler J, Mohty M, Niederwieser D, Or R, et al. Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. N Engl J Med. 2020;382:180010.
Article PubMed Google Scholar
Zeiser R, Polverelli N, Ram R, Hashmi SK, Chakraverty R, Middeke JM, et al. Ruxolitinib for glucocorticoid-refractory chronic graft-versus-host disease. N Engl J Med. 2021;385:22838.
Article CAS PubMed Google Scholar
Zeiser R, Burchert A, Lengerke C, Verbeek M, Maas-Bauer K, Metzelder SK, et al. Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia. 2015;29:20628.
Article CAS PubMed PubMed Central Google Scholar
Krger N, Shahnaz Syed Abd Kadir S, Zabelina T, Badbaran A, Christopeit M, Ayuk F. et al. Peritransplantation ruxolitinib prevents acute graft-versus-host disease in patients with myelofibrosis undergoing allogenic stem cell transplantation. Biol Blood Marrow Transplant. 2018;24:21526.
Article PubMed Google Scholar
Gooptu M, Antin JH. GVHD prophylaxis 2020. Front Immunol. 2021;12:605726.
Article CAS PubMed PubMed Central Google Scholar
Borowitz MJ, Craig FE, Digiuseppe JA, Illingworth AJ, Rosse W, Sutherland DR, et al. Guidelines for the diagnosis and monitoring of paroxysmal nocturnal hemoglobinuria and related disorders by flow cytometry. Cytom B Clin Cytom. 2010;78:21130.
Article Google Scholar
Donohue RE, Marcogliese AN, Sasa GS, Elghetany MT, Redkar AA, Bertuch AA, et al. Standardized high-sensitivity flow cytometry testing for paroxysmal nocturnal hemoglobinuria in children with acquired bone marrow failure disorders: a single center US study. Cytom B Clin Cytom. 2018;94:699704.
Article CAS Google Scholar
Zhang Y, Huo J, Liu L, Shen Y, Chen J, Zhang T, et al. Comparison of hematopoietic stem cell transplantation outcomes using matched sibling donors, haploidentical donors, and immunosuppressive therapy for patients with acquired aplastic anemia. Front Immunol. 2022;13:837335.
Article CAS PubMed PubMed Central Google Scholar
Han TT, Xu LP, Liu DH, Liu KY, Wang FR, Wang Y, et al. Recombinant human thrombopoietin promotes platelet engraftment after haploidentical hematopoietic stem cell transplantation: a prospective randomized controlled trial. Ann Hematol. 2015;94:11728.
Article CAS PubMed Google Scholar
Tang B, Huang L, Liu H, Cheng S, Song K, Zhang X, et al. Recombinant human thrombopoietin promotes platelet engraftment after umbilical cord blood transplantation. Blood Adv. 2020;4:382939.
Article CAS PubMed PubMed Central Google Scholar
Chinese Society of Hematology, Chinese Medical Association. [Chinese expert consensus on the management of hemorrhagic complications after hematopoietic stem cell transplantation(2021)]. Zhonghua Xue Ye Xue Za Zhi. 2021;42:27680.
Wang J, Zhou M, Xu JY, Zhou RF, Chen B, Wan Y. Comparison of antifungal prophylaxis drugs in patients with hematological disease or undergoing hematopoietic stem cell transplantation: a systematic review and network meta-analysis. JAMA Netw Open. 2020;3:e2017652.
Article PubMed PubMed Central Google Scholar
Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J, et al. 1994 Consensus Conference on Acute GVHD Grading. Bone Marrow Transplant. 1995;15:8258.
CAS PubMed Google Scholar
Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, Lee SJ, et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005;11:94556.
Article PubMed Google Scholar
Champlin RE, Horowitz MM, van Bekkum DW, Camitta BM, Elfenbein GE, Gale RP, et al. Graft failure following bone marrow transplantation for severe aplastic anemia: risk factors and treatment results. Blood. 1989;73:60613.
Article CAS PubMed Google Scholar
Lin F, Han T, Zhang Y, Cheng Y, Xu Z, Mo X, et al. The incidence, outcomes, and risk factors of secondary poor graft function in haploidentical hematopoietic stem cell transplantation for acquired aplastic anemia. Front Immunol. 2022;13:896034.
Article CAS PubMed PubMed Central Google Scholar
McLornan DP, Hernandez-Boluda JC, Czerw T, Cross N, Joachim Deeg H, Ditschkowski M, et al. Allogeneic haematopoietic cell transplantation for myelofibrosis: proposed definitions and management strategies for graft failure, poor graft function and relapse: best practice recommendations of the EBMT Chronic Malignancies Working Party. Leukemia. 2021;35:244559.
Article PubMed Google Scholar
Li J, Wang Y, Zhang Y, Zhang X, Pang A, Yang D, et al. Haematopoietic stem cell transplantation for hepatitis-associated aplastic anaemia and non-hepatitis-associated aplastic anaemia: a propensity score-matched analysis. Br J Haematol. 2023;201:117991.
Article PubMed Google Scholar
Aggarwal N, Manley AL, Chen J, Groarke EM, Feng X, Young NS. Effects of ruxolitinib on murine regulatory T cells are immune-context dependent. Exp Hematol. 2023;125126:169.
Article PubMed Google Scholar
Xu ZL, Huang XJ. Optimizing outcomes for haploidentical hematopoietic stem cell transplantation in severe aplastic anemia with intensive GVHD prophylaxis: a review of current findings. Expert Rev Hematol. 2021;14:44955.
Article CAS PubMed Google Scholar
Williams L, Cirrone F, Cole K, Abdul-Hay M, Luznik L, Al-Homsi AS. Post-transplantation cyclophosphamide: from HLA-haploidentical to matched-related and matched-unrelated donor blood and marrow transplantation. Front Immunol. 2020;11:636.
Article CAS PubMed PubMed Central Google Scholar
Bourgeois AL, Jullien M, Garnier A, Peterlin P, Bn MC, Guillaume T, et al. Post-transplant cyclophosphamide as sole GHVD prophylaxis after matched reduced-intensity conditioning allotransplant. Clin Transl Med. 2023;13:e1242.
Article CAS PubMed PubMed Central Google Scholar
Prata PH, Eikema DJ, Afansyev B, Bosman P, Smiers F, Diez-Martin JL, et al. Haploidentical transplantation and posttransplant cyclophosphamide for treating aplastic anemia patients: a report from the EBMT Severe Aplastic Anemia Working Party. Bone Marrow Transplant. 2020;55:10508.
Article CAS PubMed Google Scholar
Chang YJ, Zhao XY, Huang XJ. Granulocyte colony-stimulating factor-primed unmanipulated haploidentical blood and marrow transplantation. Front Immunol. 2019;10:2516.
Article CAS PubMed PubMed Central Google Scholar
Yao D, Tian Y, Li J, Li B, Lu J, Ling J, et al. Association between haploidentical hematopoietic stem cell transplantation combined with an umbilical cord blood unit and graft-versus-host disease in pediatric patients with acquired severe aplastic anemia. Ther Adv Hematol. 2022;13:20406207221134409.
Article CAS PubMed PubMed Central Google Scholar
Xu ZL, Huang XJ. Haploidentical transplants with a G-CSF/ATG-based protocol: experience from China. Blood Rev. 2023;62:101035.
Article CAS PubMed Google Scholar
Xu LP, Lu DP, Wu DP, Jiang EL, Liu DH, Huang H, et al. Hematopoietic stem cell transplantation activity in China 20202021 during the SARS-CoV-2 pandemic: a report from the Chinese Blood and Marrow Transplantation Registry Group. Transplant Cell Ther. 2023;29:136 e1e7.
Article PubMed Google Scholar
Yoshimi A, Baldomero H, Horowitz M, Szer J, Niederwieser D, Gratwohl A, et al. Global use of peripheral blood vs bone marrow as source of stem cells for allogeneic transplantation in patients with bone marrow failure. JAMA. 2016;315:198200.
Article PubMed Google Scholar
Kennedy GA, Varelias A, Vuckovic S, Le Texier L, Gartlan KH, Zhang P, et al. Addition of interleukin-6 inhibition with tocilizumab to standard graft-versus-host disease prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial. Lancet Oncol. 2014;15:14519.
Article CAS PubMed Google Scholar
Chen YB, Shah NN, Renteria AS, Cutler C, Jansson J, Akbari M, et al. Vedolizumab for prevention of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Blood Adv. 2019;3:413646.
Article CAS PubMed PubMed Central Google Scholar
Lin Y, Gu Q, Lu S, Pan Z, Yang Z, Li Y, et al. Ruxolitinib improves hematopoietic regeneration by restoring mesenchymal stromal cell function in acute graft-versus-host disease. J Clin Investig. 2023;133:e162201.
Ryu DB, Lim JY, Kim TW, Shin S, Lee SE, Park G, et al. Preclinical evaluation of JAK1/2 inhibition by ruxolitinib in a murine model of chronic graft-versus-host disease. Exp Hematol. 2021;98:3646.e2.
Article CAS PubMed Google Scholar
Hlsdnker J, Ottmller KJ, Neeff HP, Koyama M, Gao Z, Thomas OS, et al. Neutrophils provide cellular communication between ileum and mesenteric lymph nodes at graft-versus-host disease onset. Blood. 2018;131:185869.
Article PubMed PubMed Central Google Scholar
Heine A, Held SA, Daecke SN, Wallner S, Yajnanarayana SP, Kurts C, et al. The JAK-inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo. Blood. 2013;122:1192202.
Article CAS PubMed Google Scholar
Sylvine P, Thomas S, Pirayeh E. Infections associated with ruxolitinib: study in the French Pharmacovigilance database. Ann Hematol. 2018;97:9134.
Article PubMed Google Scholar
Abedin S, McKenna E, Chhabra S, Pasquini M, Shah NN, Jerkins J, et al. Efficacy, toxicity, and infectious complications in ruxolitinib-treated patients with corticosteroid-refractory graft-versus-host disease after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2019;25:168994.
Article CAS PubMed Google Scholar
Hong X, Chen Y, Lu J, Lu Q. Addition of ruxolitinib in graft-versus-host disease prophylaxis for pediatric -Thalassemia major patients after allogeneic stem cell transplantation: a retrospective cohort study. Pediatr Transplant. 2023;27:e14466.
Article CAS PubMed Google Scholar
Zhang B, Chen L, Zhou J, Zu Y, Gui R, Li Z, et al. Ruxolitinib early administration reduces acute GVHD after alternative donor hematopoietic stem cell transplantation in acute leukemia. Sci Rep. 2021;11:8501.
Article CAS PubMed PubMed Central Google Scholar
Zhao Y, Shi J, Luo Y, Gao F, Tan Y, Lai X, et al. Calcineurin inhibitors replacement by ruxolitinib as graft-versus-host disease prophylaxis for patients after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2020;26:e128e33.
Article CAS PubMed Google Scholar
Morozova EV, Barabanshikova MV, Moiseev IS, Shakirova AI, Barhatov IM, Ushal IE, et al. A prospective pilot study of graft-versus-host disease prophylaxis with post-transplantation cyclophosphamide and ruxolitinib in patients with myelofibrosis. Acta Haematol. 2021;144:15865.
Article CAS PubMed Google Scholar
See the rest here:
Addition of ruxolitinib to standard graft-versus-host disease prophylaxis for allogeneic stem cell transplantation in ... - Nature.com
Alzheimer’s Could Be Transmitted via Bone Marrow Transplants, Researchers Say – PEOPLE
A new study found that Alzheimers could potentially be accelerated by bone marrow transplants from donors with the disease.
On Thursday, a group of researchers from the University of British Columbia in Canada published a study in the scientific journal Stem Cell Reports that found that lab mice that received bone marrow transplants from other mice that had a protein associated with Alzheimer's experienced rapid cognitive decline.
The study, researchers said, could help scientists start to pinpoint what exactly contributes to Alzheimer's and other types of cognitive decline especially because so much is unknown about Alzheimer's, including whether it's caused by the brain or by genetic or environmental factors.
"This supports the idea that Alzheimers is a systemic disease where amyloids that are expressed outside of the brain contribute to central nervous system pathology," Wilfred Jefferies, an immunologist and an author on the study, told Neuroscience News.
As we continue to explore this mechanism, Alzheimers disease may be the tip of the iceberg and we need to have far better controls and screening of the donors used in blood, organ and tissue transplants as well as in the transfers of human-derived stem cells or blood products," Jefferies added.
In the study, the researchers used both healthy mice and mice showing signs of Alzheimer's disease, and transplanted bone marrow to them from mice with a hereditary form of Alzheimer's. The healthy mice began developing signs of Alzheimer's at 9 months old, and the mice that already had the Alzheimer's protein began to experience cognitive decline at 6 months old.
"The fact that we could see significant behavioral differences and cognitive decline in the APP-knockouts at 6 months was surprising but also intriguing because it just showed the appearance of the disease that was being accelerated after being transferred," the study's first author, Chaahat Singh, told Neuroscience News.
Never miss a story sign up for PEOPLE's free daily newsletter to stay up-to-date on the best of what PEOPLE has to offer, from celebrity news to compelling human interest stories.
The University of British Columbia researchers wrote that bone marrow and other medical donors should be screened for Alzheimer's before they are able to give a transfusion.
However, several other scientists who spoke with Medscape warned that the risk of a human receiving Alzheimer's via a bone marrow or stem cell transfer is very low.
Paul Morgan, a dementia researcher at Cardiff University in the United Kingdom, told the scientific outlet that the study involved a "very specific experimental situation," and that it is a "gargantuan leap" to say that there is a significant risk in humans for spreading Alzheimer's through transplants.
Go here to see the original:
Alzheimer's Could Be Transmitted via Bone Marrow Transplants, Researchers Say - PEOPLE
Microplastics dampen the self-renewal of hematopoietic stem cells by disrupting the gut microbiota-hypoxanthine-Wnt … – Nature.com
Mice
C57BL/6J (CD45.2) and C57BL6.SJL (CD45.1) mice were purchased from The Jackson Laboratory and housed under specific pathogen-free conditions. Male and female mice from 8 to 12 weeks were used in experiments and provided with a suitable environment and sufficient water and food. After a week of acclimatization, each mouse was randomly divided into groups, given 100L pure water, 0.01mg/100L, or 0.1mg/100L MPs by oral gavage every two days for five weeks in a gavage experiment (n=5 for each group). For the intravenous injection experiment, MPs were administered into mouse blood via the tail vein at a rate of 0.1g/100L per week for a duration of 4 weeks (n=5 for each group). All animal experiments were first approved by the Laboratory Animal Welfare and Ethics Committee of Zhejiang University (AP CODE: ZJU20220108).
Indocyanine green polystyrene (ICG-PS), polystyrene (PS) and polymethyl methacrylate (PMMA) particles were obtained from Suzhou Mylife Advanced Material Technology Company (China). Polyethylene (PE) particles were purchased from Cospheric (USA). Scanning electron microscopy (SEM, Nova Nano 450, FEI) was used to characterize the primary sizes and shapes of different MPs20. MPs were dispersed in ultrapure water with sonication before dynamic light scattering analysis (Zetasizer, Malvern, UK) to determine the hydrodynamic sizes and zeta potentials49.
Mice were sacrificed and organs were removed within six hours of ICG-PS gavage, including the heart, lung, kidney, spleen, liver, gastrointestinal tissues and bone marrow. Feces were collected 1h before the mice were sacrificed. Both organs and feces were monitored by ex vivo bioluminescence imaging with a small-animal imaging system50 (IVIS Spectrum, PerkinElmer).
For flow cytometry analysis and isolation of hematopoietic stem and progenitor cells, cells were stained with relevant antibodies51 in PBS with 2% fetal bovine serum for 3045min on ice. Antibody clones that were used: Sca-1-PE-Cy7, c-Kit-APC, CD150-PE, CD48-BV421, CD45.1-FITC, CD45.2 PE-Cy5, Gr-1-PE-Cy5, Mac1-PE-Cy5, IgM-PE-Cy5, CD3-PE-Cy5, CD4- PE-Cy5, CD8-PE-Cy5, CD45R-PE-Cy5 and Ter-119-PE-Cy5. Detailed antibody information is summarized in Supplementary Table S6. HSPCs were stained with a lineage antibody cocktail (Gr-1, Mac1, CD3, CD4, CD8, CD45R, TER119 and B220), Sca-1, c-Kit, CD150 and CD48. Cell types were defined as followed: LSK compartment (LinSca-1+c-Kit+), LT-HSC (LSK CD150+CD48), ST-HSC (LSK CD150CD48), MPP2 (LSK CD150+CD48+) and MPP3/4 (LSK CD150CD48+). B cells (CD45.2+Mac1Gr-1+B220+), T cells (CD45.2+Mac1Gr-1+CD3+) and myeloid cells (CD45.2+Mac1+Gr-1). Samples were analyzed on a flow cytometer (CytoFLEX LX, Beckman). For sorting HSCs, lineage antibody cocktail-conjugated paramagnetic microbeads and MACS separation columns (Miltenyi Biotec) were used to enrich Lin cells before sorting. Stained cells were re-suspended in PBS with 2% FBS and sorted directly using the Beckman moflo Astrios EQ (Beckman). Flow cytometry data were analyzed by FlowJo (BD) software.
Apoptosis of cells was detected by Annexin V staining (Yeason, China). After being extracted from the bone marrow of mice, 5106 cells were labeled with different surface markers for 30 to 45min at 4C and then twice rinsed with PBS. Subsequently, the cells were reconstituted in binding buffer and supplemented with Annexin V. After 30min of incubation, flow cytometry was detected in the FITC channel. Cell cycle analysis was performed with the fluorescein Ki-67 set (BD Pharmingen, USA), following the directions provided by the manufacturer. Briefly, a total of 5106 bone marrow cells were labeled with corresponding antibodies, as previously stated. Afterward, the cells were pre-treated with a fixation/permeabilization concentrate (Invitrogen, USA) at 4C overnight and subsequently rinsed with the binding buffer. The cells were stained with Ki-67 antibody for 1h in the dark and then with DAPI (Invitrogen) for another 5min at room temperature. Flow cytometry data were collected by a flow cytometer (CytoFLEX LX, Beckman, USA).
HSCs were sorted by flow cytometry according to the experimental group (ctrl and PSH mice, Rikenellaceae treatment or hypoxanthine treatment). 150 HSCs were seeded in triplicate on methylcellulose media52 (M3434, Stemcell Technologies, Inc.). After 8 days, the number of colonies was counted by microscopy. In addition, 5000 BM cells were seeded and analyzed the same way as HSCs. The cell culture media was diluted in PBS and subjected to centrifugation at 400g for 5min to determine the total cell number.
Recipient mice (CD45.1) were administered drinking water with Baytril (250mg/L) for 7 days pre-transplant and 10 days post-transplant. The day before transplantation, recipients received a lethal dose of radiation (4.5Gy at a time, divided into two times with an interval of 4h). In primary transplantation, 2105 bone marrow cells from the ctrl or PS group (CD45.2) mice and 2105 recipient-type (CD45.1) bone marrow cells were transplanted into recipient mice (CD45.1) mice. Cells were injected into recipients via tail vein injection. Donor chimerism was tracked using peripheral blood cells every 4 weeks for at least 16 weeks after transplantation. For secondary transplantation, donor BM cells were collected from primary transplant recipients sacrificed at 16 weeks after transplantation and transplanted at a dosage of 2106 cells into irradiated secondary recipient mice (9Gy). Analysis of donor chimerism and the cycle of transplantation in secondary transplantation were the same as in primary transplantation.
For limiting dilution assays52, 1104, 5104 and 2105 donor-derived bone marrow cells were collected from ctrl or PS mice (CD45.2) and transplanted into irradiated (9Gy) CD45.1 recipient mice with 2105 recipient-type (CD45.1) bone-marrow cells. Limiting dilution analysis was performed using ELDA software53. 16 weeks after transplantation, recipient mice with more than 1% peripheral-blood multilineage chimerism were defined as positive engraftment. On the other hand, recipient mice undergoing transplantation that had died before 16 weeks post transplantation were likewise evaluated as having failed engraftment54.
For histological analysis, small intestines were collected and fixed in 4% paraformaldehyde and embedded in paraffin, sectioned (5m thickness), and stained with H&E at ZJU Animal Histopathology Core Facility (China). We used Chius scores33,34 to evaluate the damage for each sample. The grade was as follows: 0, normal mucosa; 1, development of subepithelial Gruenhagens space at the tip of villus; 2, extension of the Gruenhagens area with moderate epithelial lifting; 3, large epithelial bulge with a few denuded villi; 4, denuded villi with lamina propria and exposed capillaries; and 5, disintegration of the lamina propria, ulceration, and hemorrhage. For TEM analysis, slices of the small intestine were fixed with 2.5% glutaraldehyde for ultra-microstructure observation of intestinal epithelial cells. The samples were postfixed for one hour at 4C with 1% osmium tetroxide and 30min with 2% uranyl acetate, followed by dehydration with a graded series of alcohol solutions (50%, 70%, 90% and 100% for 15min each) and acetone (100% twice for 20min). Subsequently, they were embedded with epon (Sigma-Aldrich, MO, US) and polymerized. Ultrathin sections (6080nm) were made, and examined using TEM (Tecnai G2 Spirit 120kV, Thermo FEI).
In the short-term and long-term mouse models for MP ingestion, mice were fasted for 4h before oral gavage of FITC-dextran (4kD, Sigma). The fluorescence intensity of FITC-dextran (50mg/100g body weight) was measured in the peripheral blood after 2h of gavage. Fluorescence was measured using a microplate reader (Molecular Devices, SpectraMax iD5) with excitation at 490nm and emission at 520 nm29.
Fecal samples (about 3050mg per sample) were collected from the ctrl, PSL and PSH mice, quickly frozen in liquid nitrogen, and stored at 80C. DNA samples for the microbial community were extracted using E.Z.N.A. Stool DNA Kit (Omega, USA), according to the manufacturers instructions. In brief, polymerase chain reaction (PCR) amplification of prokaryotic 16S rDNA gene V3V4 region was performed using the forward primer 341F (5-CCTACGGGNGGCWGCAG-3) and the reverse primer 805R (5-GACTACHVGGGTATCTAATCC-3)55. After 35 cycles of PCR, sequencing adapters and barcodes were included to facilitate amplification. The PCR products were detected by 1.5% agarose gel electrophoresis and were further purified using AMPure XT beads (Beckman Coulter Genomics, Danvers, MA, USA), while the target fragments were recovered using the AxyPrep PCR Cleanup Kit (Axygen, USA). In addition, the amplicon library was quantified with the Library Quantification Kit for Illumina (Kapa Biosciences, Woburn, MA, USA), and sequenced on the Illumina NovaSeq PE250 platform. In bioinformatics pipeline29,56, the assignment of paired-end reads to samples was determined by their unique barcode, and subsequently shortened by cutting off the barcode and primer sequence. The paired-end reads were combined by FLASH (v1.2.8). Quality filtering on the raw reads was carried out under precise parameters to obtain high-quality clean tags according to fqtrim (v0.94). The chimeric sequences were filtered by Vsearch software (v2.3.4). After the dereplication process using DADA2, we acquired a feature table and feature sequence. The bacterial sequence fragments obtained were grouped into Operational Taxonomic Units (OTUs) and compared to the Greengenes microbial gene database using QIIME2. Alpha diversity and beta diversity were generated by QIIME2, and pictures were drawn by R (v3.2.0). The species annotation sequence alignment was performed by Blast, with the SILVA and NT-16S databases as the alignment references. Additional sequencing results are provided in Supplementary Table S1. The experiment was supported by Lc-Bio Technologies.
The methods for the analysis of feces from HSCT donors were slightly different from those used for mice. All samples were stored in the GUHE Flora Storage buffer (GUHE Laboratories, China). The bacterial genomic DNA was extracted with the GHFDE100 DNA isolation kit (GUHE Laboratories, China) and quantified using a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, USA). The V4 region of the bacterial 16S rDNA genes was amplified by PCR, with the forward primer 515F (5-GTGCCAGCMGCCGCGGTAA-3) and the reverse primer 806R (5-GGACTACHVGGGTWTCTAAT-3). PCR amplicons were purified with Agencourt AMPure XP Beads (Beckman Coulter, IN) and quantified by the PicoGreen dsDNA Assay Kit (Invitrogen, USA). Following the previously reported steps57, the paired-end 2150bp sequencing was performed on the Illumina NovaSeq6000 platform. The details of bacterial OTUs are summarized in Supplementary Table S5. Sequence data analyses were performed using QIIME2 and R packages (v3.2.0).
For metabolite evaluation, samples from mice feces were prepared and detected as previously described55,58,59. In a nutshell, metabolites were extracted from feces through precooled 50% methanol buffer and stored at 80C before the LCMS analysis. All chromatographic separations were conducted using an ultra-performance liquid chromatography (UPLC) system (SCIEX, UK). A reversed phase separation was performed using an ACQUITY UPLC T3 column (100mm * 2.1mm, 1.8m, Waters, UK). The temperature of the column oven was maintained at 35C and the flow rate was 0.4mL/min. Both positive (the ionspray voltage floating set at 5000V) and negative ion modes (4500V) were analyzed using a TripleTOF 5600 Plus high-resolution tandem mass spectrometer (SCIEX, UK). The mass spectrometry data were obtained in Interactive Disassembler Professional (IDA) mode, with a time-of-flight (TOF) mass range of 60 to 1200Da. The survey scans were acquired in 150 milliseconds and product ion scans with a charge state of 1+ and 100 counts per second (counts/s) were recorded up to 12. Cycle duration was 0.56s. Stringent quality assurance (QA) and quality control (QC) procedures were applied, as the mass accuracy was calibrated every 20 samples and a QC sample was obtained every 10 samples. LCMS raw data files underwent processing in XCMS (Scripps, La Jolla, CA) to perform peak picking, peak alignment, gap filling, and sample normalization. Online KEGG was adopted to annotate metabolites through the matching between the precise molecular mass data (m/z) of samples and those from the database. PCA and volcano plot were utilized to identify ion characteristics that exhibit significant differences between the groups. The details of metabolomes can be found in Supplementary Table S2. The experiment was supported by Lc-Bio Technologies.
Before FMT, SPF mice received a 200L antibiotic treatment (1g/L ampicillin, 0.5g/L neomycin, 0.5g/L vancomycin and 1g/L metronidazole) for three consecutive days by oral gavage. Fresh feces were collected from ctrl or PS mice and resuspended in reduced PBS (0.5g/L cysteine and 0.2g/L Na2S in PBS) at a ratio of about 120mg feces/mL reduced PBS. Feces were then centrifuged at 500g for 1min to remove insolubilize particles25. Recipients (C57BL/6J mice) were administered 100mL of the supernatant from different groups by oral gavage twice every week for 4 weeks. 2 days after the last FMT, recipients were euthanized to analyze the changes in the hematopoietic system.
The Rikenellaceae strain (ATCC BAA-1961), purchased from ATCC, was cultured in an anaerobic chamber using BD Difco Dehydrated Culture Media: Reinforced Clostridial Medium at a temperature of 37C with a gas mixture of 80% N2 and 20% CO2. The final concentration of Rikenellaceae was 2108 viable c.f.u. per 100L and hypoxanthine (200mg/kg, Sigma, Germany) was dissolved in double distilled water29. Mice first received antibiotic treatment (same as FMT) and were then treated by oral gavage with 100L of either Rikenellaceae or hypoxanthine suspension three times a week for 4 weeks. Reinforced Clostridial Medium or double distilled water was used as a vehicle control, respectively. 2 days after the last administration, recipients were euthanized to analyze the changes in the hematopoietic system. To examine the impact of hypoxanthine on HSCs, we exposed bone marrow cells to direct co-culture with hypoxanthine at a concentration of 100pg/mL for a period of 3 days.
Mouse bone marrow cells were harvested by flushing the mices tibia and femur in phosphate buffered saline (PBS) with 2% fetal bovine serum (GIBCO). Harvested cells were grown into 96-well u-bottom plates containing freshly made HSC culture medium (StemSpanTM SFEM, Stemcell Tec.) with SCF (50ng/mL; PeproTech) and TPO (50ng/mL; PeproTech), at 37C with 5% CO2. For HSC culture, the medium was changed every 3 days by manually removing half of the conditioned medium and replacing it with fresh medium60. To assess the effects of WNT10A, IL-17, TNF and NF-kappa B on hematopoiesis, we cultured HSCs in a basic medium and supplemented them with related proteins (10ng/mL; Cosmo Bio, USA) or PBS as a control for two days, followed by flow cytometry analysis. Different concentrations of PS were added to the medium and tested using CCK-8 and FACS to detect the effect of MPs on cultured HSCs.
1104 HSCs were obtained in triplicate from mouse bone marrow cells from the ctrl or PSH group by flow cytometry sorting and RNA was extracted with RNAiso Plus (Takara, Japan) according to the manufacturers protocol. The concentration and integrity of RNA were examined by Qubit 2.0 and Agilent 2100 (Novogene, China), respectively. Oligo (dT)-coated magnetic beads (Novogene, China) were used to enrich eukaryotic mRNA. After cDNA synthesis and PCR amplification, the PCR product was purified using AMPure XP beads (Novogene, China) to obtain the final library. The Illumina high-throughput sequencing platform NovaSeq 6000 was used for sequencing. Analysis of gene expression was calculated by R or the DESeq2 package61. Detailed information regarding RNA-seq is listed in Supplementary Table S3.
For RNA expression analysis, total RNA from bone marrow cells was extracted using Trizol (Invitrogen, US) and resuspended in nuclease-free water. Reverse transcription was performed using the QuantiTect Reverse Transcription kit (Qiagen NV). qPCR was conducted using cDNA, primers and SYBR-green (Takara, Japan) in 20L using the ABI 7500 Q-PCR system62. Results were calculated using the RQ value (RQ=2Ct). Mouse Actin was chosen as the normalization control. Gene-specific primer sequences are shown in Supplementary Table S7.
Bone marrow and Rikenellaceae supernatant in different groups were obtained by centrifugation. Fecal supernatant was obtained from human samples. Hypoxanthine (LANSO, China) and WNT10A (EIAab, China) were measured by ELISA with respective kits according to the manufacturers protocols.
Human feces and peripheral blood samples were obtained from 14 subjects who provided grafts for HSCT patients. They were divided into graft success group and graft failure (GS)/poor graft function (GF/PGF) group, with 7 participants in each group. Research involving humans was approved by the Clinical Research Ethics Committee of the First Affiliated Hospital, College of Medicine, Zhejiang University (IIT20230067B). All participants read and signed the informed consent. Detailed information on patients was listed in Supplementary Table S4.
The Agilent 8700 Laser Direct Infrared Imaging system was utilized for fast and automated analysis of MPs in feces received from donors. An excessive nitric acid concentration (68%) was added to the sample and heated to dissolve the protein. Large particles were first intercepted with a large aperture filter and then filtered by vacuum extraction. After rinsing with ultra-pure water and ethanol several times, the materials, including MPs, were dispersed in the ethanol solution. The LDIR test was carried out when the ethanol was completely volatilized63. The sample of MPs was positioned on the standard sample stage. The stage was then put into the sample stage, and the Agilent Clarity was initiated to advance the sample stage into the sample chamber. The software rapidly scanned the chosen test area using a constant wave number of 1800cm1, and accurately detected and pinpointed the particles within the selected area. The unoccupied area devoid of particles was automatically designated as the background. The background spectrum was gathered and readjusted, followed by the visualization of detected particles and the collection of the whole infrared spectrum. After obtaining the particle spectrum, the spectrum library was utilized to carry out qualitative analysis automatically, including the inclusion picture, size, and area of each particle. The test was supported by Shanghai WEIPU Testing Technology Group.
MPs in peripheral blood from donors were tested by Py-GC/MS. Nitric acid was added to samples for digestion at 110C for 12h, and then used deionized water to make the solution weakly acidic. After concentration, the solution was dribbled into the sampling crucible of Py-GCMS and tested when the solvent in the crucible was completely volatilized17. Various standards of MPs were prepared and analyzed using Py-GCMS in order to construct the quantitative curve. PY-3030D Frontier was employed for lysis, with a lysis temperature set at 550 C. The chromatographic column dimensions were 30m in length, 0.25mm inner diameter, and 0.25m film thickness. The sample was subjected to a heat preservation period of 2min at 40C, followed by a gradual increase in temperature at a rate of around 20C per minute until it reached 320C. The sample was maintained at this temperature for 14min and the entire process takes a total of 30min. The carrier gas utilized was helium, with the ion source temperature of 230C. The split ratio employed was 5:1, and the m/z scan range spanned from 40 to 60064. The experiment was supported by Shanghai WEIPU Testing Technology Group.
Each animal experiment was tested using at least 56 replicates and each in vitro experiment was at least three replicates. Specific replication details are provided in relevant figure captions. Statistical significance was ascertained through unpaired two-tailed t-tests by GraphPad Prism when the P value was less than 0.05. Error bars in all figures indicate the standard deviation (SD).
Hereditary Alzheimer’s Transmitted Via Bone Marrow Transplants – Neuroscience News
Summary: Alzheimers disease, traditionally seen as a brain-centric condition, may have systemic origins and can be accelerated through bone marrow transplants from donors with familial Alzheimers to healthy mice.
A new study underscores the diseases potential transmission via cellular therapies and suggests screening donors for Alzheimers markers to prevent inadvertent disease transfer.
By demonstrating that amyloid proteins from peripheral sources can induce Alzheimers in the central nervous system, this research shifts the understanding of Alzheimers towards a more systemic perspective, highlighting the need for cautious screening in transplants and blood transfusions.
Key Facts:
Source: Cell Press
Familial Alzheimers disease can be transferred via bone marrow transplant, researchers show March 28 in the journalStem Cell Reports. When the team transplanted bone marrow stem cells from mice carrying a hereditary version of Alzheimers disease into normal lab mice, the recipients developed Alzheimers diseaseand at an accelerated rate.
The study highlights the role of amyloid that originates outside of the brain in the development of Alzheimers disease, which changes the paradigm of Alzheimers from being a disease that is exclusively produced in the brain to a more systemic disease.
Based on their findings, the researchers say that donors of blood, tissue, organ, and stem cells should be screened for Alzheimers disease to prevent its inadvertent transfer during blood product transfusions and cellular therapies.
This supports the idea that Alzheimers is a systemic disease where amyloids that are expressed outside of the brain contribute to central nervous system pathology, says senior author and immunologist Wilfred Jefferies, of the University of British Columbia.
As we continue to explore this mechanism, Alzheimers disease may be the tip of the iceberg and we need to have far better controls and screening of the donors used in blood, organ and tissue transplants as well as in the transfers of human derived stem cells or blood products.
To test whether a peripheral source of amyloid could contribute to the development of Alzheimers in the brain, the researchers transplanted bone marrow containing stem cells from mice carrying a familial version of the diseasea variant of the human amyloid precursor protein (APP) gene, which, when cleaved, misfolded and aggregated, forms the amyloid plaques that are a hallmark of Alzheimers disease.
They performed transplants into two different strains of recipient mice: APP-knockout mice that lacked an APP gene altogether, and mice that carried a normal APP gene.
In this model of heritable Alzheimers disease, mice usually begin developing plaques at 9 to 10 months of age, and behavioral signs of cognitive decline begin to appear at 11 to 12 months of age. Surprisingly, the transplant recipients began showing symptoms of cognitive decline much earlierat 6 months post-transplant for the APP-knockout mice and at 9 months for the normal mice.
The fact that we could see significant behavioral differences and cognitive decline in the APP-knockouts at 6 months was surprising but also intriguing because it just showed the appearance of the disease that was being accelerated after being transferred, says first author Chaahat Singh of the University of British Columbia.
In mice, signs of cognitive decline present as an absence of normal fear and a loss of short and long-term memory. Both groups of recipient mice also showed clear molecular and cellular hallmarks of Alzheimers disease, including leaky blood-brain barriers and buildup of amyloid in the brain.
Observing the transfer of disease in APP-knockout mice that lacked an APP gene altogether, the team concluded that the mutated gene in the donor cells can cause the disease and observing that recipient animals that carried a normal APP gene are susceptible to the disease suggests that the disease can be transferred to health individuals.
Because the transplanted stem cells were hematopoietic cells, meaning that they could develop into blood and immune cells but not neurons, the researchers demonstration of amyloid in the brains of APP knockout mice shows definitively that Alzheimers disease can result from amyloid that is produced outside of the central nervous system.
Finally the source of the disease in mice is a human APP gene demonstrating the mutated human gene can transfer the disease in a different species.
In future studies, the researchers plan to test whether transplanting tissues from normal mice to mice with familial Alzheimers could mitigate the disease and to test whether the disease is also transferable via other types of transplants or transfusions and to expand the investigation of the transfer of disease between species.
In this study, we examined bone marrow and stem cells transplantation. However, next it will be important to examine if inadvertent transmission of disease takes place during the application of other forms of cellular therapies, as well as to directly examine the transfer of disease from contaminated sources, independent from cellular mechanisms, says Jefferies.
Funding:
This research was supported by the Canadian Institutes of Health Research, the W. Garfield Weston Foundation/Weston Brain Institute, the Centre for Blood Research, the University of British Columbia, the Austrian Academy of Science, and the Sullivan Urology Foundation at Vancouver General Hospital.
Author: Kristopher Benke Source: Cell Reports Contact: Kristopher Benke Cell Reports Image: The image is credited to Neuroscience News
Original Research: The findings will appear in Stem Cell Reports
More here:
Hereditary Alzheimer's Transmitted Via Bone Marrow Transplants - Neuroscience News
ATG or post-transplant cyclophosphamide to prevent GVHD in matched unrelated stem cell transplantation? | Leukemia – Nature.com
Patient characteristics
The baseline characteristics of the study population are presented in Table1. A total of 8764 patients were included, from which 7725 (88%) received rATG, and 1039 (12%) received PTCy as GVHD prophylaxis.
Overall, the majority of patients were transplanted for acute leukemia (58%), myelodysplastic syndrome (MDS) (19.7%), myeloproliferative neoplasm (MPN) (9.7%) or lymphoma (9%). A high proportion of patients had a low/intermediate Disease Risk Index (DRI, 72.1%), and myeloablative conditioning (MAC) was more frequently performed (53.3%) than reduced intensity conditioning (RIC).
Patients in the rATG group were older, with a median age of 58.6 years (IQR (48.1, 65.4)) vs. 53 years in the PTCy group (IQR 38.6, 62.3) (p<0.01), with a similar proportion of males (57.3% in rATG vs. 58.9% in PTCy, p=0.33), along with a significantly lower use of TBI (14.5% vs. 24.7%, p<0.01) and lower use of MAC (52% vs. 62.3%, p<0.01). Also, the disease relapse index was lower and the year of transplant was more recent in the PTCy group (Table1). The remaining parameters were balanced between the two groups. Median follow up was 2.1 years in both arms. More detailed information is given in Table1.
Univariate outcomes are shown in Figs.1, 2and Table2. The results of the multivariate analyses are summarized in Table3. The P-values and hazard ratios (HR) presented in the following results section are derived from the multivariate analysis.
A NRM; B Overall survival, C Relapse incidence, D Progression-free survival and E GVHD-free relapse-free survival. Cumulative incidences are shown.
A Acute GVHD grades IIIV; B Acute GVHD grades IIIIV, C Chronic GVHD all grades and D Extensive chronic GVHD - Cumulative incidences are shown.
Patients receiving PTCy had a significantly lower NRM as compared to patients receiving rATG (2y incidence: 12.4% vs. 16.1%; HR: 0.72 [95% CI 0.550.94], p=0.016). Similarly, OS and PFS showed a statistically significant and clinically meaningful benefit for PTCy arm, with a higher OS (2y incidence: 73.9% vs. 65.1%; HR: 0.82 [95% CI 0.720.92], p=0.001), and a higher PFS (2y incidence: 64.9% vs. 57.2%; HR: 0.83 [95% CI 0.740.93], p<0.001). RI was lower in the PTCy arm (2y incidence: 22.8% vs. 26.6%; HR: 0.87 [95% CI 0.751.00], p=0.046).
The causes of death are given in Table4. No major differences between the two groups were apparent. Relapse of the underlying malignancy was the most frequent cause of death, accounting for ~50% of total deaths in both arms, followed by NRM causes: infections ~18%, GVHD~16% and other alloSCT-related causes ~8% of total deaths. Secondary malignancies contributed to approximately 1% of total deaths.
Overall chronic GVHD was lower in the PTCy group (2y incidence: PTCy 28.4% vs. rATG 31.4%; HR: 0.77 [95% CI 0.630.95], p=0.012). Extensive chronic GVHD was also reduced in patients receiving PTCy vs. rATG: (2y incidence: 11.9% vs. 13.5%; HR: 0.75 [95% CI 0.620.91], p=0.004).
The incidence of acute GVHD grades II-IV in patients receiving PTCy, compared to those receiving ATG was not statistically significant: (100d incidence: 24.1% vs. 26.5%; HR: 0.85 [95% CI 0.691.04], p=0.11). Similarly, for severe acute GVHD grades IIIIV (100d incidence: 8.7% vs. 9.7%; HR: 0.76 [95% CI 0.551.05], p=0.091).
GRFS was significantly higher in the PTCy arm compared to the rATG arm (2y incidence: 51% vs. 45%; HR: 0.86 [95% CI 0.750.99], p=0.035).
The EBMT Database does not contain data on graft failure/rejection. To get insight into the initial grafts success and any subsequent requirement for additional transplantation procedures, we investigated neutrophil recovery after the first alloSCT as well as the incidence of a second alloSCT. The median incidence of neutrophil recovery at days +30 and +60 in the ATG vs. PTCy groups was: d+30 ATG 96% (IC95% 95.596.4) vs. PTCy 91% (8992.7) and d+60 ATG 97.9% (97.698.2) vs. PTCy 97.4% (96.298.3). The median incidence of a second alloSCT at 2 years was 4.3% (3.84.8) in the ATG group and 3.2% (2.24.6) in the PTCy group.
Continue reading here:
ATG or post-transplant cyclophosphamide to prevent GVHD in matched unrelated stem cell transplantation? | Leukemia - Nature.com