91 results on '"Leslie S. Kean"'
Search Results
2. Blood and Marrow Transplant Clinical Trials Network State of the Science Symposium 2021: Looking Forward as the Network Celebrates its 20th Year
- Author
-
Frederick L. Locke, Bronwen E. Shaw, Mary M. Horowitz, Eric S. Leifer, Leslie S. Kean, Mary Eapen, Brent R. Logan, Edward A. Stadtmauer, Betty K. Hamilton, Yi-Bin Chen, Amy Foley, Mehdi Hamadani, Richard T. Maziarz, Karen K. Ballen, Mark C. Walters, Steven M. Devine, Marcelo C. Pasquini, John E. Levine, Marcie L. Riches, Helen E. Heslop, Eneida R. Nemecek, Richard J. Jones, Parameswaran Hari, Rachel Phelan, and Amy E. DeZern
- Subjects
medicine.medical_specialty ,Clinical Trials and Supportive Activities ,MEDLINE ,Transplants ,Disease ,Clinical Research ,medicine ,Immunology and Allergy ,Humans ,State of the science ,Bone Marrow Transplantation ,Clinical Trials as Topic ,Transplantation ,business.industry ,BMT ,Clinical study design ,Hematopoietic Stem Cell Transplantation ,Cell Therapy ,Cell Biology ,Hematology ,Stem Cell Research ,Clinical Trial ,Clinical trial ,Bone transplantation ,Family medicine ,Molecular Medicine ,business - Abstract
In 2021 the BMT CTN held the 4th State of the Science Symposium where the deliberations of 11 committees concerning major topics pertinent to a particular disease, modality, or complication of transplant, as well as two committees to consider clinical trial design and inclusion, diversity, and access as cross-cutting themes were reviewed. This article summarizes the individual committee reports and their recommendations on the highest priority questions in hematopoietic stem cell transplant and cell therapy to address in multicenter trials.
- Published
- 2021
3. Concerted changes in the pediatric single-cell intestinal ecosystem before and after anti-TNF blockade
- Author
-
Hengqi Betty Zheng, Benjamin A. Doran, Kyle Kimler, Alison Yu, Victor Tkachev, Veronika Niederlova, Kayla Cribbin, Ryan Fleming, Brandi Bratrude, Kayla Betz, Lorenzo Cagnin, Connor McGuckin, Paula Keskula, Alexandre Albanese, Maria Sacta, Joshua de Sousa Casal, Ruben van Esch, Andrew C. Kwong, Conner Kummerlowe, Faith Taliaferro, Nathalie Fiaschi, Baijun Kou, Sandra Coetzee, Sumreen Jalal, Yoko Yabe, Michael Dobosz, Matthew F. Wipperman, Sara Hamon, George D. Kalliolias, Andrea Hooper, Wei Keat Lim, Sokol Haxhinasto, Yi Wei, Madeline Ford, Lusine Ambartsumyan, David L. Suskind, Dale Lee, Gail Deutsch, Xuemei Deng, Lauren V. Collen, Vanessa Mitsialis, Scott B. Snapper, Ghassan Wahbeh, Alex K. Shalek, Jose Ordovas-Montanes, and Leslie S. Kean
- Subjects
Cell type ,Leukocyte migration ,Stromal cell ,Myeloid ,business.industry ,Disease ,medicine.disease ,Bioinformatics ,Inflammatory bowel disease ,Immune system ,medicine.anatomical_structure ,medicine ,Vector (molecular biology) ,business - Abstract
Crohn’s disease is an inflammatory bowel disease (IBD) which most often presents with patchy lesions in the terminal ileum and colon and requires complex clinical care. Recent advances in the targeting of cytokines and leukocyte migration have greatly advanced treatment options, but most patients still relapse and inevitably progress. Although single-cell approaches are transforming our ability to understand the barrier tissue biology of inflammatory disease, comprehensive single-cell RNA-sequencing (scRNA-seq) atlases of IBD to date have largely sampled pre-treated patients with established disease. This has limited our understanding of which cell types, subsets, and states at diagnosis are predictive of disease severity and response to treatment. Here, through a combined clinical, flow cytometric, and scRNA-seq study, we profile diagnostic human biopsies from the terminal ileum of treatment-naive pediatric patients with Crohn’s disease (pediCD; n=14) and from non-inflamed pediatric controls with functional gastrointestinal disorders (FGID; n=13). To fully resolve and annotate epithelial, stromal, and immune cell states among the 201,883 single-cell transcriptomes, we develop and deploy a principled and unbiased tiered clustering approach, ARBOL, yielding 138 FGID and 305 pediCD end cell clusters. Notably, through both flow cytometry and scRNA-seq, we observe that at the level of broad cell types, treatment-naive pediCD is not readily distinguishable from FGID in cellular composition. However, by integrating high-resolution scRNA-seq analysis, we identify significant differences in cell states that arise during pediCD relative to FGID. Furthermore, by closely linking our scRNA-seq analysis with clinical meta-data, we resolve a vector of lymphoid, myeloid, and epithelial cell states in treatment-naive samples which can distinguish patients with less severe disease (those not on anti-TNF therapies (NOA)), from those with more severe disease at presentation who require anti-TNF therapies. Moreover, this vector was also able to distinguish those patients that achieve a full response (FR) to anti-TNF blockade from those more treatment-resistant patients who only achieve a partial response (PR). Our study jointly leverages a treatment-naive cohort, high-resolution principled scRNA-seq data analysis, and clinical outcomes to understand which baseline cell states may predict inflammatory disease trajectory.
- Published
- 2021
- Full Text
- View/download PDF
4. Myelopathy Due to CAR-T Related Neurotoxicity Treated with Siltuximab
- Author
-
Leslie S. Kean, Yasmin Aghajan, Henrikas Vaitkevicius, Austin I. Kim, Alison Yu, Caron A. Jacobson, and Matthew Robertson
- Subjects
medicine.medical_specialty ,business.industry ,Refractory Hematologic Malignancy ,Encephalopathy ,Neurotoxicity ,medicine.disease ,Gastroenterology ,Siltuximab ,Myelopathy ,chemistry.chemical_compound ,Cytokine release syndrome ,chemistry ,Internal medicine ,Toxicity ,medicine ,Neurology (clinical) ,business ,Stroke - Abstract
Chimeric antigen receptor T (CART) cell therapy is highly effective for relapsed/refractory hematologic malignancy [1,2]; however, cytokine release syndrome (CRS) and neurotoxicity are observed in up to 77% of patients [3]. In large case series, the most common presentations of neurotoxicity were encephalopathy (57%), headache (42%), tremor (38%) and aphasia (35%). CART mediated spinal cord toxicity is not well characterized. Structural neurologic damage (stroke and intracranial hemorrhage) was only observed in 1-2% and seizures were seen in 1%-8% of cases [3, 4]. Neuroimaging findings in patients with neurotoxicity are rare and not specific.
- Published
- 2021
- Full Text
- View/download PDF
5. Phase II Trial of Costimulation Blockade With Abatacept for Prevention of Acute GVHD
- Author
-
Yvonne Suessmuth, Shalini Shenoy, Catherine Bresee, Alison Yu, Amelia Langston, Scott N. Furlan, Andrew C. Harris, Steven E. Bosinger, Muna Qayed, Sungjin Kim, Brandi Bratrude, Maxim Norkin, Audrey G. Tumlin, Jeffrey H. Davis, James Rhodes, John T. Horan, Courtney McCracken, Urvi Kapoor, Alexandria Narayan, Leslie S. Kean, Kayla Betz, Benjamin Watkins, John E. Levine, Bruce R. Blazar, Aleksandra Petrovic, Sung Won Choi, Kayla Cribbin, Michael Grimley, James L.M. Ferrara, Scott Gillespie, Kyle Hebert, Ted Gooley, Marcelo C. Pasquini, Shauna Sinclair, Michael A. Pulsipher, Mourad Tighiouart, Roger Giller, David A. Jacobsohn, Nahal R. Lalefar, Kirk R. Schultz, Christine Duncan, Edmund K. Waller, Gregory A. Yanik, Victor Tkachev, Andre Rogatko, and Nosha Farhadfar
- Subjects
Oncology ,musculoskeletal diseases ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Graft vs Host Disease ,Disease ,Tacrolimus ,law.invention ,Abatacept ,Young Adult ,Randomized controlled trial ,law ,immune system diseases ,Internal medicine ,medicine ,Humans ,Young adult ,Child ,Cause of death ,Aged ,Costimulation blockade ,business.industry ,High mortality ,Hematopoietic Stem Cell Transplantation ,ORIGINAL REPORTS ,Middle Aged ,Clinical trial ,surgical procedures, operative ,Methotrexate ,Cyclosporine ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
PURPOSE Severe (grade 3-4) acute graft-versus-host disease (AGVHD) is a major cause of death after unrelated-donor (URD) hematopoietic cell transplant (HCT), resulting in particularly high mortality after HLA-mismatched transplantation. There are no approved agents for AGVHD prevention, underscoring the critical unmet need for novel therapeutics. ABA2 was a phase II trial to rigorously assess safety, efficacy, and immunologic effects of adding T-cell costimulation blockade with abatacept to calcineurin inhibitor (CNI)/methotrexate (MTX)-based GVHD prophylaxis, to test whether abatacept could decrease AGVHD. METHODS ABA2 enrolled adults and children with hematologic malignancies under two strata: a randomized, double-blind, placebo-controlled stratum (8/8-HLA-matched URD), comparing CNI/MTX plus abatacept with CNI/MTX plus placebo, and a single-arm stratum (7/8-HLA-mismatched URD) comparing CNI/MTX plus abatacept versus CNI/MTX CIBMTR controls. The primary end point was day +100 grade 3-4 AGVHD, with day +180 severe-AGVHD-free-survival (SGFS) a key secondary end point. Sample sizes were calculated using a higher type-1 error (0.2) as recommended for phase II trials, and were based on predicting that abatacept would reduce grade 3-4 AGVHD from 20% to 10% (8/8s) and 30% to 10% (7/8s). ABA2 enrolled 142 recipients (8/8s, median follow-up = 716 days) and 43 recipients (7/8s, median follow-up = 708 days). RESULTS In 8/8s, grade 3-4 AGVHD was 6.8% (abatacept) versus 14.8% (placebo) ( P = .13, hazard ratio = 0.45). SGFS was 93.2% (CNI/MTX plus abatacept) versus 82% (CNI/MTX plus placebo, P = .05). In the smaller 7/8 cohort, grade 3-4 AGVHD was 2.3% (CNI/MTX plus abatacept, intention-to-treat population), which compared favorably with a nonrandomized matched cohort of CNI/MTX (30.2%, P < .001), and the SGFS was better (97.7% v 58.7%, P < .001). Immunologic analysis revealed control of T-cell activation in abatacept-treated patients. CONCLUSION Adding abatacept to URD HCT was safe, reduced AGVHD, and improved SGFS. These results suggest that abatacept may substantially improve AGVHD-related transplant outcomes, with a particularly beneficial impact on HLA-mismatched HCT.
- Published
- 2021
6. End the pain: Start with antiracism
- Author
-
Natasha M. Archer, Leslie S. Kean, and Tolulope O Rosanwo
- Subjects
Adult ,Male ,medicine.medical_specialty ,Anemia ,Extramural ,business.industry ,Attitude of Health Personnel ,media_common.quotation_subject ,MEDLINE ,Pain ,Hematology ,Anemia, Sickle Cell ,medicine.disease ,Racism ,Family medicine ,medicine ,Humans ,business ,media_common - Published
- 2020
7. Isolation of a Highly Purified HSC-enriched CD34+CD90+CD45RA− Cell Subset for Allogeneic Transplantation in the Nonhuman Primate Large-animal Model
- Author
-
Hans-Peter Kiem, Michelle Hoffman, Lucrezia Colonna, Anai M. Perez, Leslie S. Kean, and Stefan Radtke
- Subjects
Transplantation ,Allogeneic transplantation ,business.industry ,medicine.medical_treatment ,lcsh:Surgery ,Hematopoietic stem cell ,lcsh:RD1-811 ,Hematopoietic stem cell transplantation ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Cancer research ,Bone Marrow and Stem Cell Transplantation ,Bone marrow ,Stem cell ,Progenitor cell ,business - Abstract
Background. Allogeneic hematopoietic stem cell transplantation (allo-HCT) is a common treatment for patients suffering from different hematological disorders. Allo-HCT in combination with hematopoietic stem cell (HSC) gene therapy is considered a promising treatment option for millions of patients with HIV+ and acute myeloid leukemia. Most currently available HSC gene therapy approaches target CD34-enriched cell fractions, a heterogeneous mix of mostly progenitor cells and only very few HSCs with long-term multilineage engraftment potential. As a consequence, gene therapy approaches are currently limited in their HSC targeting efficiency, very expensive consuming huge quantities of modifying reagents, and can lead to unwanted side effects in nontarget cells. We have previously shown that purified CD34+CD90+CD45RA− cells are enriched for multipotent HSCs with long-term multilineage engraftment potential, which can reconstitute the entire hematopoietic system in an autologous nonhuman primate transplant model. Here, we tested the feasibility of transplantation with purified CD34+CD90+CD45RA− cells in the allogeneic setting in a nonhuman primate model. Methods. To evaluate the feasibility of this approach, CD34+CD90+CD45RA− cells from 2 fully major histocompatibility complex-matched, full sibling rhesus macaques were sort-purified, quality controlled, and transplanted. Engraftment and donor chimerism were evaluated in the peripheral blood and bone marrow of both animals. Results. Despite limited survival due to infectious complications, we show that the large-scale sort-purification and transplantation of CD34+CD90+CD45RA− cells is technically feasible and leads to rapid engraftment of cells in bone marrow in the allogeneic setting and absence of cotransferred T cells. Conclusions. We show that purification of an HSC-enriched CD34+ subset can serve as a potential stem cell source for allo-HCTs. Most importantly, the combination of allo-HCT and HSC gene therapy has the potential to treat a wide array of hematologic and nonhematologic disorders.
- Published
- 2020
8. Defining success with cellular therapeutics: the current landscape for clinical end point and toxicity analysis
- Author
-
Leslie S. Kean
- Subjects
0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Bioinformatics ,Immunotherapy, Adoptive ,Biochemistry ,03 medical and health sciences ,medicine ,Animals ,Humans ,Therapeutic window ,End point ,Extramural ,business.industry ,Review Series ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Hematologic Diseases ,030104 developmental biology ,medicine.anatomical_structure ,Hematologic disease ,Hematologic Neoplasms ,business - Abstract
Cellular therapies play a major and expanding role in the treatment of hematologic diseases. For each of these therapies, a narrow therapeutic window exists, where efficacy is maximized and toxicities minimized. This review focuses on one of the most established cellular therapies, hematopoietic stem cell transplant, and one of the newest cellular therapies, chimeric antigen receptor–T cells. In this review, I will discuss the current state of the field for clinical end point analysis with each of these therapeutics, including their critical toxicities, and focus on the major elements of success for each of these complex treatments for hematologic disease.
- Published
- 2018
- Full Text
- View/download PDF
9. Reprint of: Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR
- Author
-
Partow Kebriaei, Miguel-Angel Perales, Leslie S. Kean, and Michel Sadelain
- Subjects
0301 basic medicine ,Transplantation ,biology ,business.industry ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,T cell ,Hematology ,Immunotherapy ,Bioinformatics ,medicine.disease ,CD19 ,Chimeric antigen receptor ,03 medical and health sciences ,Cytokine release syndrome ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Intensive care ,medicine ,biology.protein ,business ,B cell - Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
- Published
- 2018
- Full Text
- View/download PDF
10. Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR
- Author
-
Partow Kebriaei, Leslie S. Kean, Michel Sadelain, and Miguel-Angel Perales
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Lymphoma ,Chronic lymphocytic leukemia ,T cell ,Antigens, CD19 ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,0302 clinical medicine ,Genome editing ,hemic and lymphatic diseases ,Intensive care ,medicine ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,B cell ,Gene Editing ,Salvage Therapy ,Transplantation ,Leukemia ,business.industry ,Genes, Transgenic, Suicide ,Hematology ,medicine.disease ,Chimeric antigen receptor ,Cytokine release syndrome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Cytokines ,Nervous System Diseases ,business - Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
- Published
- 2018
- Full Text
- View/download PDF
11. MI-Immune/1801: Lessons from an Ongoing, Multi-Center Trial Involving Biospecimen Collection for Prospective Microbiome and Immune Profiling in Patients Undergoing Reduced Intensity Conditioning Allogeneic HCT
- Author
-
Wael Saber, Andrew R. Rezvani, Stephanie Waldvogel, Suman R. Das, Mehdi Hamadani, Alan Howard, Mary M. Horowitz, Mary Riwes, Ashley Spahn, Leslie S. Kean, Miguel-Angel Perales, Hany Elmariah, Shernan G. Holtan, Merav Bar, Brent R. Logan, Hemant S. Murthy, Javier Bolaños-Meade, Ami S. Bhatt, Armin Rashidi, John E. Levine, Monzr M. Al Malki, Mahasweta Gooptu, Robert R. Jenq, Brandi Bratrude, Marcie L. Riches, Karamjeet S. Sandhu, Anthony D. Sung, Erin F. Brooks, William B. Clark, Richard J. Jones, Saurabh Chhabra, and Lyndsey Runaas
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Allogeneic hct ,Cell Biology ,Hematology ,Biochemistry ,Biospecimen Collection ,Immune profiling ,Immune system ,Internal medicine ,Reduced Intensity Conditioning ,medicine ,In patient ,Microbiome ,business - Abstract
Introduction: The gut microbiome is a potentially modifiable factor in treatment related outcomes in allogeneic hematopoietic cell transplant (HCT). Prior studies have linked pre- or mid-treatment gut microbiome diversity with risk for treatment related morbidity and mortality. However, these studies have been limited by the inclusion of one or only a few institutions and the lack of longitudinal sampling with high quality metadata. These limitations complicate the interpretation of microbiome alterations over the course of HCT. Methods: To overcome these, we devised and implemented a large-scale biospecimen collection protocol in conjunction with BMT CTN 1703, a randomized, multicenter, Phase III trial of tacrolimus/methotrexate vs. post-transplant cyclophosphamide/tacrolimus/mycophenolate mofetil in reduced intensity conditioning (RIC) allogeneic HCT (NCT03959241). Patients enrolled on 1703 were optionally co-enrolled in the companion immune and microbiome profiling study, 1801 (MI-IMMUNE). This involved blood, urine and stool sampling before conditioning (PCON), then weekly starting on day 0 through day 77 (through day 84 for blood), and on days 98, 180, 270, 365 and 730. For all enrolled participants where the donor consented, residual donor cells were saved from the empty hematopoietic stem cell product bag for later analysis. Additionally, the protocol included one-time blood, urine and stool sample collection from consented matched related donors (MRDs) prior to stem cell collection. Starting with protocol version 4.0 on February 1, 2021, participation in 1801 stool collection was required for the first six out of eighteen sample collection timepoints. Participation in later stool and urine timepoint collections remained optional. Here we review the feasibility of creating a multi-institutional biobank. Additionally, we assess the success of our strategies by calculating sample collection compliance and standard deviation in compliance across centers for each timepoint. Results: On June 18, 2021, BMT CTN 1703/1801 closed to accrual with 431 patients enrolled on 1703; 323 patients from 36 centers were co-enrolled on 1801. 304 (94%) provided study samples, making this the largest prospective microbiome and immune profiling study in HCT patients to date. As of July 6, 2021, 3,683 blood, 2,668 urine, and 2,098 stool samples had been collected. Across the first 6 timepoints for all participating centers, blood, urine and stool sample collection averaged 93%, 82%, and 74% compliance, respectively. Of the 99 (30%) patients enrolled on 1801 with a MRD, 34 (34%) donors consented to sample collection. Sample collection compliance was lower for MRDs than for patients on the study with 76%, 74%, and 62% of expected blood, urine and stool samples collected, respectively, from this group. For stool collection exclusively, a median of 5 samples were collected per patient across the first 6 timepoints (median of 6 across all timepoints) with 93 (31%) of patients completing a full sample set through Day 28. 139 (46%) patients provided at least one sample after day 28; these represented 37% of the total samples collected to date. The PCON sample, which provides an important measure of pre-treatment gut microbiome diversity, had the third highest compliance with 74% of patients providing a sample. Surprisingly, Day 28 had the lowest compliance (66%) and highest standard deviation (37%) possibly because this timepoint often falls around the time of hospital discharge. Between PCON and day 28, the standard deviation between sites in the average collection compliance (24%) and number of samples collected per patient (1.1) was small indicating the successful adoption of stool collection across institutions. Table 1 summarizes sample collection statistics. Conclusion: Overall this study has resulted in a large, novel biobank of blood, urine and stool samples from patients undergoing RIC allogeneic HCT at 36 centers across the US. This will serve as a valuable resource for investigating the role of the gut microbiome in long term health outcomes following HCT. Although the results of 1801 are forthcoming given ongoing sample collection, the size and composition of the biobank to date clearly demonstrate the feasibility of implementing multi-institutional stool collection. This study represents a critical step towards the large-scale adoption of microbiome sampling as a diagnostic tool. Figure 1 Figure 1. Disclosures Chhabra: GSK: Honoraria. Clark: Kadmon: Consultancy. Horowitz: Mesoblast: Research Funding; Shire: Research Funding; Vertex: Research Funding; Stemcyte: Research Funding; Vor Biopharma: Research Funding; Janssen: Research Funding; Miltenyi Biotech: Research Funding; Kiadis: Research Funding; Sobi: Research Funding; Kite/Gilead: Research Funding; Pfizer, Inc: Research Funding; Jazz Pharmaceuticals: Research Funding; Magenta: Consultancy, Research Funding; Medac: Research Funding; Novartis: Research Funding; GlaxoSmithKline: Research Funding; Daiicho Sankyo: Research Funding; Xenikos: Research Funding; Omeros: Research Funding; Orca Biosystems: Research Funding; Pharmacyclics: Research Funding; Regeneron: Research Funding; Tscan: Research Funding; Takeda: Research Funding; CSL Behring: Research Funding; Genentech: Research Funding; Gamida Cell: Research Funding; Chimerix: Research Funding; Bristol-Myers Squibb: Research Funding; bluebird bio: Research Funding; Astellas: Research Funding; Amgen: Research Funding; Allovir: Consultancy; Actinium: Research Funding; Sanofi: Research Funding; Seattle Genetics: Research Funding. Jenq: Microbiome DX: Consultancy; Merck: Consultancy; Prolacta: Consultancy, Membership on an entity's Board of Directors or advisory committees; Kaleido: Consultancy, Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Research Funding; Seres: Consultancy, Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; LisCure: Consultancy, Membership on an entity's Board of Directors or advisory committees; MaaT Pharma: Consultancy, Membership on an entity's Board of Directors or advisory committees; Karius: Consultancy. Levine: Equillium Bio: Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy, Research Funding; Kamada: Research Funding; Biogen: Research Funding; Omeros: Membership on an entity's Board of Directors or advisory committees; Symbio: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Talaris Therapeutics: Membership on an entity's Board of Directors or advisory committees; Viracor: Patents & Royalties: GVHD biomarker patent with royalties from Viracor; Mesoblast: Consultancy, Research Funding; X4 Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Murthy: CRISPR Therapeutics: Research Funding. Riches: ATARA Biotherapeutics: Other: Payment; BioIntelect: Membership on an entity's Board of Directors or advisory committees; Jazz Pharmaceuticals: Other: Payment. Sung: Merck: Research Funding; Novartis: Research Funding; Enterome: Research Funding; Seres: Research Funding; AVROBIO: Consultancy; Abbott Nutrition: Honoraria; Clasado: Other: Research Product; DSM: Other: Research Product. Al Malki: Neximmune: Consultancy; Jazz Pharmaceuticals, Inc.: Consultancy; Rigel Pharma: Consultancy; Hansa Biopharma: Consultancy; CareDx: Consultancy. Rezvani: Kaleido: Other: One-time scientific advisory board; Nohla Therapeutics: Other: One-time scientific advisory board; Pharmacyclics-Abbvie: Research Funding; US Department of Justice: Consultancy. Bolaños-Meade: Incyte Corp: Consultancy. Holtan: Incyte: Consultancy, Research Funding; Generon: Consultancy. Saber: Govt. COI: Other. Hamadani: Sanofi, Genzyme, AstraZeneca, BeiGene: Speakers Bureau; Janssen, Incyte, ADC Therapeutics, Omeros, Morphosys, Kite: Consultancy; Takeda, Spectrum Pharmaceuticals and Astellas Pharma: Research Funding. Kean: Bluebird Bio: Research Funding; Bristol Myers Squibb: Patents & Royalties: From clinical trial data, Research Funding; Vertex: Consultancy; Novartis: Consultancy; Gilead: Research Funding; Regeneron: Research Funding; EMD Serono: Consultancy. Perales: Cidara: Honoraria; Servier: Honoraria; Incyte: Honoraria, Other; Equilium: Honoraria; Takeda: Honoraria; Novartis: Honoraria, Other; Nektar Therapeutics: Honoraria, Other; NexImmune: Honoraria; MorphoSys: Honoraria; Omeros: Honoraria; Karyopharm: Honoraria; Sellas Life Sciences: Honoraria; Merck: Honoraria; Miltenyi Biotec: Honoraria, Other; Medigene: Honoraria; Bristol-Myers Squibb: Honoraria; Kite/Gilead: Honoraria, Other; Celgene: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
12. Low Rates of Venoocclusive Disease and Non-Relapse Mortality in Pediatric Patients Undergoing Allogeneic Hematopoietic Cell Transplantation Following Inotuzumab Ozogamicin Reinduction for Relapsed or Refractory B Cell Acute Lymphoblastic Leukemia
- Author
-
Steven P. Margossian, John T. Horan, Lewis B. Silverman, Donna Neuberg, Malika Kapadia, Susanne H.C. Baumeister, Jonathan Paolino, Jiemin Yang, Leslie Lehmann, Melissa A. Burns, Francesca Alvarez Calderon, Andrew E. Place, Jennifer Whangbo, Christine Duncan, and Leslie S. Kean
- Subjects
Inotuzumab ozogamicin ,Oncology ,medicine.medical_specialty ,Hematopoietic cell ,business.industry ,Immunology ,Venoocclusive disease ,Cell Biology ,Hematology ,B-cell acute lymphoblastic leukemia ,Biochemistry ,Transplantation ,Refractory ,Internal medicine ,Medicine ,Nonrelapse mortality ,business ,medicine.drug - Abstract
Background: Inotuzumab ozogamicin (INO) is a humanized, anti-CD22 antibody conjugated to calicheamicin with FDA approval for treatment of adults with relapsed or refractory (r/r) B-acute lymphoblastic leukemia (B-ALL). While it is associated with favorable complete remission rates in r/r B-ALL patients (pts), hepatotoxicity both during treatment and with subsequent hematopoietic cell transplantation (HCT) remains a challenge. In general, venoocclusive disease (VOD) during HCT in pediatric pts is estimated at 10-20%. Following treatment with INO, however, up to 52% of pts undergoing HCT have been reported to develop VOD, of which 18% of cases were fatal (Bhojwani et al. 2019). Here we report our institutional HCT experience in pediatric pts with r/r B-ALL who underwent HCT following INO reinduction and our strategy of using defibrotide prophylaxis to reduce the risk of VOD. Methods: A retrospective chart review was conducted for pts with r/r B-ALL who underwent HCT at our institution from December 2015 to January 2021. Clinical characteristics, laboratory parameters, and radiographic evaluations were reviewed. Patients who received ≥1 course of INO during reinduction were included in the INO group. Development of VOD was defined as a clinical diagnosis of VOD post-HCT based on either the Baltimore, Seattle, or European Society for Blood and Marrow Transplantation (EBMT) diagnostic criteria.Clinical characteristics were summarized. Time to VOD, graft versus host disease (GVHD), non-relapse mortality (NRM), and relapse were estimated under the competing risk setting, in which death without the event of interest was considered to be an absorbing competing risk. Results: Fifty-seven patients with relapsed or refractory B-ALL were transplanted between December 2015 and January 2021. Seventeen (30%) received at least 1 course (3 doses) of INO as part of their pre-HCT reinduction regimen. Fifty-five of 57 pts (97%), including all pts in the INO-group received a myeloablative regimen (51 total body irradiation/cyclophosphamide-based, 4 busulfan-based). Two patients in the non-INO group received a reduced-intensity regimen. Median time from last dose of INO to HCT (Day 0) was 34 days (range 24-71 days). Median number of INO doses received was 6 (range 3-9). Four patients had been transplanted previously, 1 in the INO group and 3 in the non-INO group. Two of these patients developed VOD during their current HCT, both from the non-INO group. Pts receiving INO were more likely to be transplanted in CR3 or higher, and to have required 3 regimens in reinduction to achieve that CR. Median follow-up was 20 months. Rates of VOD were similar between the INO (3 of 17, 18%) and non-INO (4 of 40, 10%) groups (p=0.4, Figure 1). All cases were severe or very severe by EBMT criteria. There was no VOD-related mortality in either group. Defibrotide prophylaxis was used in 82% (14 of 17) of pts in the INO group. Of the 3 patients in the INO group who developed VOD, 2 received defibrotide prophylaxis and 1 did not. Reasons to defer defibrotide prophylaxis included increased risk of CNS bleeding and provider preference. Defibrotide prophylaxis was used in 15% of pts in the non-INO group (6 of 40). In the non-INO group, 1 of 4 patients who developed VOD had received defibrotide prophylaxis. All pts received vitamin E and ursodiol per institutional practice. There were no treatment-related deaths in the INO group and no statistically significant differences were detected in (NRM) (0% vs 10%, p=0.25), median time to engraftment (24 vs 26.5 days, p=0.1), or GVHD (24% vs 55%, p=0.1) in the INO vs non-INO groups. Conclusions: HCT was well tolerated with low rates of VOD and NRM in pediatric pts following reinduction with INO. The results of ongoing prospective studies will provide further insight into the role of INO for reinduction of r/r B-ALL. In our cohort, rates of VOD were similar between pts who had and had not previously received INO, with the majority of pts in the INO group receiving defibrotide prophylaxis during HCT, suggesting that inotuzumab ozogamicin reinduction may safely be used with this approach. Figure 1 Figure 1. Disclosures Margossian: Cue Biopharma: Current Employment. Silverman: Takeda, Servier, Syndax, Jazz Pharmaceuticals: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees. Neuberg: Pharmacyclics: Research Funding; Madrigal Pharmaceuticals: Other: Stock ownership. OffLabel Disclosure: This study evaluates the safety of transplant following inotuzumab ozogamicin re-induction for children with B lymphoblastic leukemia.
- Published
- 2021
- Full Text
- View/download PDF
13. Improved Overall Survival of Patients Treated with Abatacept in Combination with a Calcineurin Inhibitor and Methotrexate Following 7/8 HLA-Matched Unrelated Allogeneic Hematopoietic Stem Cell Transplantation: Analysis of the Center for International Blood and Marrow Transplant Research Database
- Author
-
Martin Polinsky, Mei-Jie Zhang, Leslie S. Kean, Marcelo C. Pasquini, Roxanne Kapikian, Andres Gomez, Sean E. Connolly, Linda J. Burns, Tzuyung Douglas Kou, Brian J. Gavin, Michael T. Hemmer, and Xiao-Ying Tang
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Abatacept ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Human leukocyte antigen ,Hematopoietic stem cell transplantation ,Biochemistry ,Calcineurin ,Bone transplantation ,Internal medicine ,Overall survival ,medicine ,Methotrexate ,Database research ,business ,medicine.drug - Abstract
Introduction: Acute graft versus host disease (aGVHD) is the most common cause of early non-relapse mortality following allogeneic hematopoietic stem cell transplantation (HSCT; McDonald-Hyman C, et al. Sci Transl Med 2015). aGVHD is caused by a series of complex interactions between donor T cells and recipient antigen-presenting cells that result in immune-mediated tissue damage in HSCT recipients. Inhibition of T-cell costimulation has been recognized as a highly effective approach to preventing T-cell allo-activation (McDonald-Hyman C, et al. Sci Transl Med 2015; Koura DT, et al. Biol Blood Marrow Transplant 2013). Abatacept, a CTLA-4-IgG1 fusion protein, binds to CD80/86 on antigen-presenting cells and inhibits CD28 costimulatory signaling required for T-cell activation (Malmstrom V, et al. Nat Rev Immunol 2017). A phase 2 study (ClinicalTrials.gov: NCT01743131) reported 73.6% (80% confidence interval [CI]: 62.0-82.2) overall survival (OS) at 2 years in recipients of a 7/8 human leukocyte antigen (HLA)-single mismatch unrelated donor (7/8 MMUD) HSCT following treatment with abatacept + a standard aGVHD prophylaxis regimen (calcineurin inhibitor [CNI] + methotrexate [MTX] without [−] antithymocyte globulin [ATG]), compared with 45.3% (80% CI: 39.3-51.1; P = 0.002) in a standard treatment cohort (CNI + MTX − ATG) of matched controls from the Center for International Blood and Marrow Transplant Research (CIBMTR; Watkins B, et al. J Clin Oncol 2021). The aim of this current real-world analysis was to further evaluate the impact of abatacept on survival of 7/8 MMUD HSCT recipients, treated with CNI + MTX − ATG with or without abatacept, from the CIBMTR database of all allogeneic HSCTs performed in the United States in recent years. Methods: In this observational study, patients (≥ 6 years of age with leukemia, lymphoma, or myelodysplastic syndrome, whose first allogenic HSCT was with a 7/8 MMUD between 2011 and 2018) were treated with CNI + MTX − ATG with or without abatacept. OS (defined as time between date of transplant and documented date of death) was evaluated at 181 days post-transplant by weighted log-rank test with inverse propensity scores, obtained using logistic regression models as weights (inverse probability of treatment weighting [IPTW]) to reduce bias due to confounding. The marginal hazard ratio (HR) and 95% CI were estimated by a weighted Cox proportional hazards model. Kaplan-Meier curves show the estimated survival probabilities over time. Exploratory analyses of OS were evaluated in 7/8 MMUD HSCT recipients treated with abatacept + CNI + MTX (without ATG) versus CNI + MTX + ATG, and versus those treated with post-transplant cyclophosphamide-based (PT-Cy) GVHD prophylaxis. Results: For the primary analysis, 216 patients (54 [25%] abatacept + CNI + MTX − ATG and 162 [75%] CNI + MTX − ATG) were included. Key patient demographics and characteristics were generally similar across treatment groups in weighted samples using IPTW (Table 1). The majority of patients were male and had performance scores of 90-100; had acute myeloid leukemia; and had received myeloablative conditioning, peripheral blood stem cell grafts, and tacrolimus. Kaplan-Meier OS rates at day 180 post-transplant by weighted log-rank test with inverse propensity scores (95% CI) were 98% (78-100) for patients treated with abatacept + CNI + MTX − ATG and 75% (67-82) for those treated with CNI + MTX − ATG (P = 0.0028). The marginal HRs (95% CI) were 0.06 (0.01-0.27) and 0.07 (0.01-0.30) using treatment only and treatment plus disease status as covariates, respectively. A Kaplan-Meier plot of estimated survival probability over time is shown in Figure 1. The exploratory analysis showed that patients treated with abatacept + CNI + MTX had improved OS HRs (95% CI) of 0.08 (0.02-0.36) versus CNI + MTX + ATG, and 0.15 (0.03-0.67) versus PT-Cy-based GVHD prophylaxis. Conclusions: Patients treated with abatacept in combination with CNI + MTX − ATG after allogeneic HSCT had significantly better OS at day 180 compared with patients treated with only CNI + MTX − ATG, a standard treatment. Results from this real-world database study are consistent with and are supportive of the findings from the phase 2 study. Study support: This study was sponsored by Bristol Myers Squibb. Professional medical writing and editorial assistance was provided by Fiona Boswell, PhD, of Caudex and was funded by Bristol Myers Squibb. Figure 1 Figure 1. Disclosures Kean: Novartis: Consultancy; Bluebird Bio: Research Funding; Gilead: Research Funding; EMD Serono: Consultancy; Regeneron: Research Funding; Bristol Myers Squibb: Patents & Royalties: From clinical trial data, Research Funding; Vertex: Consultancy. Burns: Kyowa Kirin International: Research Funding; Medac GmbH: Research Funding; OncoImmune: Research Funding; Sanofi: Research Funding; Fate: Research Funding; PrioThera: Research Funding; Bristol Meyers Squibb: Research Funding; Astellas Pharma Inc.: Research Funding. Kou: Bristol Myers Squibb: Current Employment. Kapikian: Bristol Myers Squibb: Current Employment. Hemmer: Pfizer: Honoraria. Connolly: Bristol Myers Squibb: Current Employment, Current holder of individual stocks in a privately-held company. Polinsky: Bristol Myers Squibb: Current Employment, Current holder of individual stocks in a privately-held company. Gavin: Bristol Myers Squibb: Current Employment, Current equity holder in publicly-traded company. Gomez: Bristol Myers Squibb: Other: I have no financial relationship with an ineligible company. I am an employee of Analysis Group, Inc. (AG), an economic consulting firm. AG receives consulting fees from ineligible companies, including BMS.. Pasquini: GlaxoSmithKline: Research Funding; Kite Pharma: Research Funding; Novartis: Research Funding; Bristol Myers Squibb: Consultancy, Research Funding. OffLabel Disclosure: Abatacept, a CTLA-4 fusion protein, is not an FDA-approved indication for prophylaxis of acute graft versus host disease
- Published
- 2021
- Full Text
- View/download PDF
14. Comparable Outcomes for Matched and Mismatched Unrelated Donor (URD) Transplantation with the Addition of Abatacept to Standard Graft Versus Host Disease Prophylaxis
- Author
-
Kayla Betz, Chao Zhang, John T. Horan, Nahal R. Lalefar, Aleksandra Petrovic, Catherine Bresee, Leslie S. Kean, Scott Gillespie, Nosha Farhadfar, Edmund K. Waller, Gregory A. Yanik, David A. Jacobsohn, Yvonne Suessmuth, Roger Giller, James Rhodes, Christine Duncan, Sungjin Kim, Mourad Tighiouart, Shalini Shenoy, Jeffrey H. Davis, Sung Choi, Muna Qayed, Mike A. Pulsipher, Benjamin Watkins, Brandi Bratrude, Courtney McCracken, Alison Yu, Kirk R. Schultz, Michael Grimley, Andre Rogatko, Scott N. Furlan, Amelia Langston, Andrew C. Harris, Bruce R. Blazar, Kayla Cribbin, and Maxim Norkin
- Subjects
Transplantation ,medicine.medical_specialty ,business.industry ,Abatacept ,Mismatched Unrelated Donor ,Cell Biology ,Hematology ,medicine.disease ,Surgery ,Graft-versus-host disease ,medicine ,Molecular Medicine ,Immunology and Allergy ,business ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
15. Immune Reconstitution Following Unrelated Donor Hematopoietic Cell Transplantation for Pediatric Non-Malignant Diseases Using Abatacept Graft-Versus-Host Disease Prophylaxis
- Author
-
Benjamin Watkins, Leslie S. Kean, John Horan, Ann E. Haight, Muna Qayed, Yvonne Suessmuth, Elizabeth Stenger, and Kuang-Yueh Chiang
- Subjects
Transplantation ,Hematopoietic cell ,business.industry ,Abatacept ,Non malignant ,Cell Biology ,Hematology ,medicine.disease ,Immune system ,Graft-versus-host disease ,Unrelated Donor ,Immunology ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
16. Predicting Immune Pathology after Hematopoietic Stem Cell Transplant (HCT) with Day 100 Transcriptomics: Naïve CD4 T Cell Expansion Versus Regulatory Programming Predicts Patients Who Develop De Novo Chronic Gvhd (CGVHD) Versus Those Displaying Operational Immune Tolerance
- Author
-
Amelia Langston, Ben Watkins, John Horan, Muna Qayed, Brandi Bratrude, Yvonne Suessmuth, Leslie S. Kean, Kayla Betz, James Kaminski, and Victor Tkachev
- Subjects
Transplantation ,Cd4 t cell ,business.industry ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Immune tolerance ,Transcriptome ,Immune system ,medicine.anatomical_structure ,Immunology ,Molecular Medicine ,Immunology and Allergy ,Chronic gvhd ,Medicine ,business - Published
- 2021
- Full Text
- View/download PDF
17. A Canine Model of Chronic Graft-versus-Host Disease
- Author
-
Andrew R. Rezvani, Michele Spector, Scott S. Graves, Bruce Swearingen, Diane Stone, Leslie S. Kean, Maura H. Parker, George E. Sale, Steven Lawrence Rosinski, and Rainer Storb
- Subjects
Transplantation Conditioning ,medicine.medical_treatment ,Graft vs Host Disease ,Bronchiolitis obliterans ,Lymphocyte Depletion ,Article ,03 medical and health sciences ,Dogs ,0302 clinical medicine ,Transplantation Immunology ,medicine ,Animals ,Transplantation, Homologous ,Bone Marrow Transplantation ,Transplantation ,biology ,business.industry ,Dog leukocyte antigen ,Graft Survival ,Immunosuppression ,Hematology ,Total body irradiation ,medicine.disease ,Histocompatibility ,Disease Models, Animal ,surgical procedures, operative ,Graft-versus-host disease ,030220 oncology & carcinogenesis ,Blood Buffy Coat ,Chronic Disease ,Immunology ,biology.protein ,Methotrexate ,Unrelated Donors ,business ,Immunosuppressive Agents ,Whole-Body Irradiation ,030215 immunology ,medicine.drug - Abstract
In long-term survivors of allogeneic hematopoietic cell transplantation (HCT), chronic graft-versus-host disease (GVHD) is the major cause of morbidity and mortality and a major determinant of quality of life. Chronic GVHD responds poorly to current immunosuppressive drugs, and while T cell depletion may be preventive, this gain is offset by increased relapse rates. A significant impediment to progress in treating chronic GVHD has been the limitations of existing animal models. The goal of this study was to develop a reproducible comprehensive model of chronic GVHD in the dog. Ten recipient dogs received 920 cGy total body irradiation, infusion of marrow, and an infusion of buffy coat cells from a dog leukocyte antigen (DLA)–mismatched unrelated donor. Postgrafting immunosuppression consisted of methotrexate (days 1, 3, 6, 11) and cyclosporine. The duration of cyclosporine administration was limited to 80 days instead of the clinically used 180 days. This was done to contain costs, as chronic GVHD was expected to develop at earlier time points. All recipients were given ursodiol for liver protection. One dog had graft failure and 9 dogs showed stable engraftment. Eight of the 9 developed de novo chronic GVHD. Dogs progressed with clinical signs of chronic GVHD over a period of 43 to 164 (median, 88) days after discontinuation of cyclosporine. Target organs showed the spectrum of chronic GVHD manifestations that are typically seen clinically. These included lichenoid changes of the skin, fasciitis, ocular involvement (xerophthalmia), conjunctivitis, bronchiolitis obliterans, salivary gland involvement, gingivitis, esophageal involvement, and hepatic involvement. Peripheral blood lymphocyte surface antigen expression of CD28 and inducible costimulator was elevated in dogs with GHVD compared with those in normal dogs, but not significantly so. Serum levels of IL-8 and monocyte chemotactic protein-1 in GVHD-affected dogs at time of euthanasia were elevated, whereas levels of IL-15 were depressed compared with those in normal dogs. Results indicate that the canine model is well suited for future studies aimed at preventing or treating chronic GVHD.
- Published
- 2017
- Full Text
- View/download PDF
18. The Knife’s Edge of Tolerance: Inducing Stable Multilineage Mixed Chimerism but With a Significant Risk of CMV Reactivation and Disease in Rhesus Macaques
- Author
-
Shan Yu, Dollnovan Tran, Kelly Hamby, Christian P. Larsen, Sanjeev Gumber, Benjamin Watkins, Allan D. Kirk, Charlotte E. Hotchkiss, Karnail Singh, Amitinder Kaur, Jennifer Lane, Andrew B. Adams, Linda C. Cendales, Bruce R. Blazar, Scott N. Furlan, Victor Tkachev, Leslie S. Kean, Katie Zeleski, and Hengqi Zheng
- Subjects
0301 basic medicine ,Transplantation Conditioning ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Cytomegalovirus ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,030230 surgery ,Communicable Diseases ,Belatacept ,Article ,Immune tolerance ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Transplantation, Homologous ,Immunology and Allergy ,Pharmacology (medical) ,Transplantation Chimera ,Transplantation ,business.industry ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Skin Transplantation ,Total body irradiation ,Macaca mulatta ,Regimen ,030104 developmental biology ,medicine.anatomical_structure ,Cytomegalovirus Infections ,Immunology ,Transplantation Tolerance ,Virus Activation ,business ,Busulfan ,medicine.drug - Abstract
Although stable mixed-hematopoietic chimerism induces robust immune tolerance to solid organ allografts in mice, the translation of this strategy to large animal models and to patients has been challenging. We have previously shown that in MHC-matched nonhuman primates (NHPs), a busulfan plus combined belatacept and anti-CD154-based regimen could induce long-lived myeloid chimerism, but without T cell chimerism. In that setting, donor chimerism was eventually rejected, and tolerance to skin allografts was not achieved. Here, we describe an adaptation of this strategy, with the addition of low-dose total body irradiation to our conditioning regimen. This strategy has successfully induced multilineage hematopoietic chimerism in MHC-matched transplants that was stable for as long as 24 months posttransplant, the entire length of analysis. High-level T cell chimerism was achieved and associated with significant donor-specific prolongation of skin graft acceptance. However, we also observed significant infectious toxicities, prominently including cytomegalovirus (CMV) reactivation and end-organ disease in the setting of functional defects in anti-CMV T cell immunity. These results underscore the significant benefits that multilineage chimerism-induction approaches may represent to transplant patients as well as the inherent risks, and they emphasize the precision with which a clinically successful regimen will need to be formulated and then validated in NHP models.
- Published
- 2017
- Full Text
- View/download PDF
19. Advances in targeting co-inhibitory and co-stimulatory pathways in transplantation settings: the Yin to the Yang of cancer immunotherapy
- Author
-
Bruce R. Blazar, Laurence A. Turka, and Leslie S. Kean
- Subjects
Graft Rejection ,0301 basic medicine ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Receptors, Antigen, T-Cell ,Context (language use) ,030230 surgery ,Lymphocyte Activation ,Article ,Immune tolerance ,Immunomodulation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Costimulatory and Inhibitory T-Cell Receptors ,Cancer immunotherapy ,Neoplasms ,Immune Tolerance ,medicine ,Animals ,Humans ,Immunology and Allergy ,business.industry ,Antibodies, Monoclonal ,Immunosuppression ,Organ Transplantation ,Immunotherapy ,Transplantation ,Tolerance induction ,030104 developmental biology ,business ,Neuroscience ,Signal Transduction - Abstract
In the past decade, the power of harnessing T cell co-signaling pathways has become increasingly understood to have significant clinical importance. In cancer immunotherapy, the field has concentrated on two related modalities: First, targeting cancer antigens through highly activated chimeric antigen T cells (CAR-Ts) and second, re-animating endogenous quiescent T cells through checkpoint blockade. In each of these strategies, the therapeutic goal is to re-ignite T cell immunity, in order to eradicate tumors. In transplantation, there is also great interest in targeting T cell co-signaling, but with the opposite goal: in this field, we seek the Yin to cancer immunotherapy’s Yang, and focus on manipulating T cell co-signaling to induce tolerance rather than activation. In this review, we discuss the major T cell signaling pathways that are being investigated for tolerance-induction, detailing pre-clinical studies and the path to the clinic for many of these molecules. These include blockade of co-stimulation pathways and agonism of coinhibitory pathways, in order to achieve the delicate state of balance that is transplant tolerance: a state which guarantees lifelong transplant acceptance without ongoing immunosuppression, and with preservation of protective immune responses. In the context of the clinical translation of immune tolerance strategies, we discuss the significant challenge that is embodied by the fact that targeted pathway modulators may have opposing effects on tolerance based on their impact on effector versus regulatory T cell biology. Achieving this delicate balance holds the key to the major challenge of transplantation: lifelong control of alloreactivity while maintaining an otherwise intact immune system.
- Published
- 2017
- Full Text
- View/download PDF
20. Predicting Immune Pathology after Hematopoietic Stem Cell Transplant with Transcriptomics: Naïve CD4 T Cell Expansion at Day 100 Predicts Patients with De Novo Chronic Gvhd
- Author
-
Amelia Langston, Yvonne Suessmuth, John T. Horan, Steven E. Bosinger, Victor Tkachev, Bruce R. Blazar, Kathryn L. Pellegrini, Muna Qayed, Kayla Betz, James Kaminski, Leslie S. Kean, Alison Yu, Ben Watkins, and Brandi Bratrude
- Subjects
Cd4 t cell ,business.industry ,Immunology ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biochemistry ,Transcriptome ,medicine.anatomical_structure ,Immune system ,immune system diseases ,hemic and lymphatic diseases ,Medicine ,Chronic gvhd ,business - Abstract
Background: Chronic graft-versus-host disease (CGVHD) is the leading cause of long-term morbidity and mortality following hematopoietic stem cell transplant (HCT) and occurs in over 50% of patients undergoing unrelated donor HCT. Despite its frequency, the mechanisms driving this disease remain incompletely understood, making its prevention and successful treatment challenging. To address this issue, we have undertaken a transcriptomic analysis of T cell reconstitution after unrelated donor HCT, to dissect mechanisms driving CGVHD. Methods: The patients studied were enrolled on a Phase 2, randomized, placebo-controlled trial of abatacept for GVHD prevention in patients receiving 8/8 unrelated-donor HCT for hematologic malignancies (NCT01743131). All immune analyses in the current study were performed on patients randomized to standard GVHD prophylaxis with calcineurin inhibition + methotrexate alone (placebo cohort, n =69), and thus provide insights into the drivers of CGVHD during standard unrelated donor HCT. On Day +100, CD4+ T cells were purified from the peripheral blood of these patients, and then analyzed by RNASeq. To determine the transcriptomic drivers of CGVHD without the confounder of significant prior acute GVHD (AGVHD) or exposure to steroids, we focused on profiling the CD4+ transcriptome of de novo CGVHD (CGVHD which develops in the absence of prior grade II-IV AGVHD, n = 7) and compared these patients to those who were 'operationally tolerant' and never developed either grade II-IV AGVHD or any CGVHD (n= 4). Gene expression from the resulting transcriptomes was quantified using kallisto. Differentially expressed (DE) genes were identified using DESeq2 (threshold for DE, adjusted (for multiple testing) p Results: DE analysis identified 101 genes that were significantly upregulated in CD4+ T cells from de novo CGVHD group and 54 genes that were significantly upregulated in the 'operationally tolerant' group (Figure 1A). GSEA identified that the mostly highly enriched signatures in patients with de novo CGVHD encompassed naïve CD4+ transcriptional programing (Figure 1B-C), in agreement with flow cytometric analysis, which also demonstrated expansion of CD4+ naïve T cells at Day +100 in patients developing de novo CGVHD compared to those demonstrating operational tolerance (Figure 1D). Importantly, the naïve CD4+ T cell signatures that were identified were distinct from those defining CD4+ stem cell memory T cells (which did not enrich in the de novo CGVHD cohort). In contrast, the gene signature of the operationally tolerant patients were enriched for regulatory gene sets (Figure 1C), consistent with a large body of evidence demonstrating that Treg expansion can be protective against CGVHD. Discussion: This study represents, to our knowledge, the first interrogation of the transcriptomic features of patients developing de novo CGVHD versus those operationally tolerant patients who develop neither significant AGVHD nor CGVHD after HCT. These patients may represent a particularly effective cohort in which to study immunologic drivers of CGVHD, given their freedom from prior treatment with corticosteroids, which can confound downstream transcriptomic analyses. Our data provide compelling evidence for a prominent naïve CD4+ T cell signature in patients who develop moderate-to-severe CGVHD despite their lack of antecedent AGVHD. These results are provocative, as they implicate a cell subset that is often considered more quiescent (naïve T cells) as associated with patients who develop immune pathology associated with CGVHD. These results suggest that naïve CD4+ T cells may represent a potent reservoir for alloreactivity, that, once activated, can cause significant disease. This would be in agreement with the implications of previously reported trials of naïve T cell depletion, which resulted in significant control of CGVHD. These results suggest that strategies to restrain naïve T cell pathogenic activation after Day +100 may improve CGVHD outcomes, and that the CD4+ T cell transcriptomic signature at this timepoint could be developed into a robust immunologic biomarker for the risk of developing CGVHD versus operational tolerance after HCT. Figure 1 Disclosures Watkins: Bristol Myers Squib: Honoraria. Qayed:Novartis: Consultancy; Mesoblast: Consultancy. Blazar:Tmunity: Other: Co-founder; KidsFirst Fund: Research Funding; BlueRock Therapeutics: Research Funding; Childrens' Cancer Research Fund: Research Funding; BlueRock Therapeuetic: Consultancy; Magenta Therapeutics: Consultancy; Fate Therapeutics Inc.: Research Funding. Horan:Bristol Myers Squib: Honoraria, Research Funding. Langston:Kadmon Corporation: Research Funding; Astellas Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Research Funding; Incyte: Research Funding; Bristol Myers Squib: Research Funding; Chimerix: Research Funding; Takeda: Research Funding. Kean:fortyseven: Consultancy; regeneron: Research Funding; hifibio: Consultancy; kymab: Consultancy; Bristol Meyers Squibb: Research Funding; gilead: Research Funding; novartis: Consultancy; bluebird bio: Research Funding; magenta: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
21. Risk factors for transplant-associated thrombotic microangiopathy and mortality in a pediatric cohort
- Author
-
Leslie Lehmann, Clement Ma, Maia Lee, Michelle Schoettler, Christine Duncan, Steven P. Margossian, Leslie S. Kean, and Pei-Chi Kao
- Subjects
Male ,medicine.medical_specialty ,Thrombotic microangiopathy ,Graft vs Host Disease ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Risk Factors ,Internal medicine ,hemic and lymphatic diseases ,Medicine ,Humans ,Child ,Retrospective Studies ,Transplantation ,business.industry ,Thrombotic Microangiopathies ,Hazard ratio ,Acute kidney injury ,Hematopoietic Stem Cell Transplantation ,Retrospective cohort study ,Hematology ,medicine.disease ,030220 oncology & carcinogenesis ,Cohort ,Female ,Complication ,business ,030215 immunology ,Tissue biopsy - Abstract
Transplant-associated thrombotic microangiopathy (TA-TMA) is a well-recognized complication of hematopoietic cell transplantation (HCT). Diagnosis is challenging and in the absence of a tissue biopsy, TA-TMA is provisionally diagnosed by meeting clinical criteria. In this study, we describe the prevalence, outcomes, and risk factors for meeting 2 different diagnostic criteria for TA-TMA and for increased transplant-related mortality (TRM). In this retrospective study of 307 pediatric HCT patients, records were reviewed for the first 100 days after HCT. Patients who were diagnosed with TA-TMA by a provider during this time were included. In addition, the Cho et al criteria (2010) and Jodele et al (2014) TA-TMA criteria were applied retrospectively. Eight patients (2.6%) were diagnosed with TA-TMA by their provider. However, on retrospective review, 20% and 36% met the Cho and Jodele criteria for TA-TMA, respectively. Overall survival was significantly worse (P < .0001) and TRM was significantly higher in patients who met criteria for TA-TMA (MC-TA-TMA) (P < .0001). After controlling for comorbid conditions, MC-TA-TMA (hazard ratio [HR], 10.9; P = .0001) and grade 3/4 acute graft-versus-host-disease (aGVHD) (HR 3.5; P = .01) remained independently associated with increased TRM. Among allogeneic HCT recipients, features associated with an increased risk for MC-TA-TMA included ≥2 HCT, concurrent grade 3/4 aGVHD and concurrent infections. Among patients who MC-TA-TMA, LDH ≥2 times the upper limit of normal (P = .001), the need for ≥2 antihypertensive medications (P < .0001), and acute kidney injury (P = .003) were associated with significantly increased TRM.
- Published
- 2019
22. A phase I study of sirolimus in combination with metronomic therapy (CHOAnome) in children with recurrent or refractory solid and brain tumors
- Author
-
Yvonne Suessmuth, Thomas Cash, Tobey J. MacDonald, Kelly C. Goldsmith, Benjamin Watkins, Muna Qayed, Howard M. Katzenstein, Leslie S. Kean, Cynthia Wetmore, Rachel Tanos, and Mourad Tighiouart
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Adolescent ,Maximum Tolerated Dose ,Malignant peripheral nerve sheath tumor ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,Glioma ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Mucositis ,Humans ,Child ,Cyclophosphamide ,Etoposide ,Medulloblastoma ,Sirolimus ,Dose-Response Relationship, Drug ,business.industry ,Brain Neoplasms ,Hematology ,medicine.disease ,Metronomic Chemotherapy ,Celecoxib ,030220 oncology & carcinogenesis ,Primitive neuroectodermal tumor ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Administration, Metronomic ,Female ,Sarcoma ,business ,030215 immunology ,Anaplastic astrocytoma - Abstract
Background/purpose To determine the maximum tolerated dose, toxicities, and response of sirolimus combined with oral metronomic therapy in pediatric patients with recurrent and refractory solid and brain tumors. Procedure Patients younger than 30 years of age with recurrent, refractory, or high-risk solid and brain tumors were eligible. Patients received six-week cycles of sirolimus with twice daily celecoxib, and alternating etoposide and cyclophosphamide every three weeks, with Bayesian dose escalation over four dose levels (NCT01331135). Results Eighteen patients were enrolled: four on dose level (DL) 1, four on DL2, eight on DL3, and two on DL4. Diagnoses included solid tumors (Ewing sarcoma, osteosarcoma, malignant peripheral nerve sheath tumor, rhabdoid tumor, retinoblastoma) and brain tumors (glioblastoma multiforme [GBM], diffuse intrinsic pontine glioma, high-grade glioma [HGG], medulloblastoma, ependymoma, anaplastic astrocytoma, low-grade infiltrative astrocytoma, primitive neuroectodermal tumor, nongerminomatous germ cell tumor]. One dose-limiting toxicity (DLT; grade 4 neutropenia) was observed on DL2, two DLTs (grade 3 abdominal pain and grade 3 mucositis) on DL3, and two DLTs (grade 3 dehydration and grade 3 mucositis) on DL4. The recommended phase II dose of sirolimus was 2 mg/m2 (DL3). Best response was stable disease (SD) in eight patients, and partial response (PR) in one patient with GBM. A patient with HGG was removed from the study with SD and developed PR without further therapy. Western blot analysis showed inhibition of phospho-S6 kinase in all patients during the first cycle of therapy. Conclusion The combination of sirolimus with metronomic chemotherapy is well tolerated in children. A phase II trial of this combination is ongoing.
- Published
- 2019
23. Potent Interaction between CMV Reactivation and Gvhd: Immunologic Evidence for Blunting of CMV-Driven Immune Reconstitution in the Setting of Gvhd
- Author
-
Muna Qayed, Benjamin Watkins, Yvonne Suessmuth, Kayla Betz, Leslie S. Kean, Amelia Langston, Brandi Bratrude, John T. Horan, and Alison Yu
- Subjects
Immune system ,business.industry ,Immunology ,Medicine ,Cell Biology ,Hematology ,Cmv reactivation ,business ,Biochemistry - Abstract
Allogeneic hematopoietic cell transplantation (HCT) may be curative for patients with marrow and immune disorders, but graft-vs-host-disease (aGVHD) and infections cause significant morbidity and non-relapse mortality. We have conducted a multicenter, double blind, placebo-controlled phase II trial of costimulation blockade with abatacept (Aba) combined with standard GVHD prophylaxis with a calcineurin inhibitor and methotrexate (CNI + MTX) following HLA matched unrelated donor transplant (n=142). In order to assess the effects of Aba on immune reconstitution, and to assess whether this reconstitution is influenced during CMV reactivation, we longitudinally evaluated post-transplant whole blood samples with multiparameter flow cytometry using markers for CD3, CD4, CD8, CD197 and CD45RA to measure reconstitution of CD4 and CD8 T cell populations and their respective memory subsets over time. Results: We observe that post-transplant CMV reactivation induces a marked expansion of CD8 effector memory (EM) cells, which is similar in magnitude for Aba vs placebo patients. We found that development of moderate (gr 2-4) or severe (gr 3-4) GVHD was not associated with an increased frequency of CMV reactivation, but patients with moderate GVHD showed a blunted expansion of CD8 EM cells compared to those without GVHD, and CD8 EM expansion was essentially absent among CMV reactivating patients with severe aGVHD. Clinical correlates will be presented. Conclusions: Our results suggest that adding abatacept to CNI/MTX does not materially affect reconstitution of T cell immunity in the presence or absence of CMV reactivation, but aGVHD remains a major driver of compromised immune recovery after HCT. Disclosures Watkins: Bristol Myers Squibb: Research Funding. Qayed:Mesoblast: Consultancy; Novartis: Consultancy. Horan:Bristol Myers Squib: Honoraria, Research Funding. Kean:gilead: Research Funding; bluebird bio: Research Funding; fortyseven: Consultancy; magenta: Research Funding; regeneron: Research Funding; hifibio: Consultancy; kymab: Consultancy; Bristol Meyers Squibb: Research Funding; novartis: Consultancy. Langston:Kadmon Corporation: Research Funding; Bristol Myers Squib: Research Funding; Incyte: Research Funding; Chimerix: Research Funding; Takeda: Research Funding; Astellas Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
24. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune Cell Signatures of Ulcerative Colitis and Crohn’s Disease
- Author
-
Liza Konnikova, Jessica Toothaker, Rima Fawaz, Jared Silverstein, Randi G. Pleskow, Michael Field, Susanna Huh, Beth-Ann Norton, Sabina Sabharwal, Amit S. Grover, Laurie N. Fishman, Peng Liu, Silvana Bonilla, Rachel W. Winter, Jose Ordovas-Montanes, Alex K. Shalek, Punyanganie S. de Silva, Anne A. Wolf, Jodie Ouahed, Bruce H. Horwitz, Sonia Arora Ballal, Victor L. Fox, Frederick L. Makrauer, Marko Vukovic, Sonia Friedman, Leslie S. Kean, Scott B. Snapper, Sarah Wall, Menno Verhave, Leslie M. Higuchi, Athos Bousvaros, Munir Mobassaleh, Stacy A. Kahn, Collin C. McCourt, Jessica R. Allegretti, Naamah L. Zitomersky, Joshua R. Korzenik, Dennis J. Spencer, Alejandro Flores, Vanessa Mitsialis, Jeff Goldsmith, Lori A. Zimmerman, George C. Tseng, Matthew J. Hamilton, Paul A. Rufo, Brian P. Regan, Tamar Parmet, and Christine K. Lee
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Colon ,Regulatory T cell ,Plasmacytoid dendritic cell ,CD38 ,Inflammatory bowel disease ,Peripheral blood mononuclear cell ,Article ,Immunophenotyping ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Crohn Disease ,medicine ,Humans ,RNA-Seq ,Intestinal Mucosa ,Child ,Immunity, Cellular ,Hepatology ,business.industry ,Innate lymphoid cell ,Gastroenterology ,Dendritic cell ,Flow Cytometry ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Case-Control Studies ,Immunology ,Colitis, Ulcerative ,Female ,030211 gastroenterology & hepatology ,Single-Cell Analysis ,business - Abstract
Background & Aims Studies are needed to determine the mechanisms of mucosal dysregulation in patients with inflammatory bowel diseases (IBDs) and differences in inflammatory responses of patients with ulcerative colitis (UC) vs Crohn’s disease (CD). We used mass cytometry (CyTOF) to characterize and compare immune cell populations in the mucosa and blood from patients with IBD and without IBD (controls) at single-cell resolution. Methods We performed CyTOF analysis of colonic mucosa samples (n = 87) and peripheral blood mononuclear cells (n = 85) from patients with active or inactive UC or CD and controls. We also performed single-cell RNA sequencing, flow cytometry, and RNA in situ hybridization analyses to validate key findings. We used random forest modeling to identify differences in signatures across subject groups. Results Compared with controls, colonic mucosa samples from patients with IBD had increased abundances of HLA-DR+CD38+ T cells, including T-regulatory cells that produce inflammatory cytokines; CXCR3+ plasmablasts; and IL1B+ macrophages and monocytes. Colonic mucosa samples from patients with UC were characterized by expansion of IL17A+ CD161+ effector memory T cells and IL17A+ T-regulatory cells; expansion of HLA-DR+CD56+ granulocytes; and reductions in type 3 innate lymphoid cells. Mucosal samples from patients with active CD were characterized by IL1B+HLA-DR+CD38+ T cells, IL1B+TNF+IFNG+ naive B cells, IL1B+ dendritic cells (DCs), and IL1B+ plasmacytoid DCs. Peripheral blood mononuclear cells from patients with active CD differed from those of active UC in that the peripheral blood mononuclear cells from patients with CD had increased IL1B+ T-regulatory cells, IL1B+ DCs and IL1B+ plasmacytoid DCs, IL1B+ monocytes, and fewer group 1 innate lymphoid cells. Random forest modeling differentiated active UC from active CD in colonic mucosa and blood samples; top discriminating features included many of the cellular populations identified above. Conclusions We used single-cell technologies to identify immune cell populations specific to mucosa and blood samples from patients with active or inactive CD and UC and controls. This information might be used to develop therapies that target specific cell populations in patients with different types of IBD.
- Published
- 2020
- Full Text
- View/download PDF
25. 317 CYTOF ANALYSIS OF HUMAN COLON AND BLOOD REVEALS DISTINCT IMMUNE SIGNATURES OF ULCERATIVE COLITIS AND CROHN'S DISEASE
- Author
-
Jessica Toothaker, Sarah Wall, Jeff Goldsmith, Alex K. Shalek, Michael Field, Peng Liu, Jose Ordovas-Montanes, Leslie S. Kean, Tamar Parmet, Liza Konnikova, Vanessa Mitsialis, Scott B. Snapper, Collin C. McCourt, and George C. Tseng
- Subjects
Crohn's disease ,Immune system ,Hepatology ,business.industry ,Immunology ,Gastroenterology ,Medicine ,business ,medicine.disease ,Ulcerative colitis ,Human colon - Published
- 2020
- Full Text
- View/download PDF
26. Human CD19-Targeted Mouse T Cells Induce B Cell Aplasia and Toxicity in Human CD19 Transgenic Mice
- Author
-
Yosef Refaeli, Jessie L. Barnum, Angela Panoskaltsis-Mortari, Jeffrey S. Miller, Heather Campbell, Zhengming Xiong, Govindarajan Thangavelu, Scott N. Furlan, Mark J. Osborn, Megan J. Riddle, Michael C. Jensen, Christopher A. Pennell, Cameron McDonald-Hyman, Bruce R. Blazar, Leslie S. Kean, Meghan D. Storlie, Michael Loschi, and Jakub Tolar
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Transgene ,T-Lymphocytes ,Antigens, CD19 ,Mice, Transgenic ,Immunotherapy, Adoptive ,CD19 ,03 medical and health sciences ,Interferon-gamma ,Mice ,Antigen ,Drug Discovery ,Genetics ,Medicine ,Animals ,Humans ,Molecular Biology ,B cell ,Pharmacology ,B-Lymphocytes ,biology ,business.industry ,Interleukin-6 ,medicine.disease ,Chimeric antigen receptor ,Cytokine release syndrome ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Commentary ,Molecular Medicine ,Original Article ,Female ,Antibody ,business - Abstract
The clinical success of chimeric antigen receptor (CAR) T cell therapy for CD19(+) B cell malignancies can be limited by acute toxicities and immunoglobulin replacement needs due to B cell aplasia from persistent CAR T cells. Life-threatening complications include cytokine release syndrome and neurologic adverse events, the exact etiologies of which are unclear. To elucidate the underlying toxicity mechanisms and test potentially safer CAR T cells, we developed a mouse model in which human CD19 (hCD19)-specific mouse CAR T cells were adoptively transferred into mice whose normal B cells express a hCD19 transgene at hemizygous levels. Compared to homozygous hCD19 transgenic mice that have ∼75% fewer circulating B cells, hemizygous mice had hCD19 frequencies and antigen density more closely simulating human B cells. Hemizygous mice given a lethal dose of hCD19 transgene-expressing lymphoma cells and treated with CAR T cells had undetectable tumor levels. Recipients experienced B cell aplasia and antigen- and dose-dependent acute toxicities mirroring patient complications. Interleukin-6 (IL-6), interferon γ (IFN-γ), and inflammatory pathway transcripts were enriched in affected tissues. As in patients, antibody-mediated neutralization of IL-6 (and IFN-γ) blunted toxicity. Apparent behavioral abnormalities associated with decreased microglial cells point to CAR-T-cell-induced neurotoxicity. This model will prove useful in testing strategies designed to improve hCD19-specific CAR T cell safety.
- Published
- 2018
27. Reduced-intensity conditioning for hematopoietic cell transplant for HLH and primary immune deficiencies
- Author
-
Mary Eapen, Leslie S. Kean, Peter Dawson, Kirk R. Schultz, Christopher Bryant, Alyssa Ramirez, James A. Connelly, Lauri Burroughs, Julie-An Talano, Michael A. Pulsipher, Catherine M. Bollard, Sung-Yun Pai, Carl E. Allen, Jeffrey R. Andolina, Elizabeth Stenger, Shalini Shenoy, Rabi Hanna, K. Scott Baker, Rebecca A. Marsh, and Philip Roehrs
- Subjects
Melphalan ,Adult ,Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,medicine.medical_treatment ,Immunology ,Graft vs Host Disease ,Antineoplastic Agents ,Hematopoietic stem cell transplantation ,Biochemistry ,Gastroenterology ,Donor lymphocyte infusion ,Lymphohistiocytosis, Hemophagocytic ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Child ,Alemtuzumab ,Survival analysis ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant ,Cell Biology ,Hematology ,medicine.disease ,Survival Analysis ,Fludarabine ,Transplantation ,surgical procedures, operative ,Graft-versus-host disease ,Treatment Outcome ,030220 oncology & carcinogenesis ,Child, Preschool ,Female ,business ,Vidarabine ,030215 immunology ,medicine.drug - Abstract
Allogeneic hematopoietic cell transplantation (HCT) with myeloablative conditioning for disorders associated with excessive inflammation such as hemophagocytic lymphohistiocytosis (HLH) is associated with early mortality. A multicenter prospective phase 2 trial of reduced-intensity conditioning with melphalan, fludarabine, and intermediate-timing alemtuzumab was conducted for HLA matched or single HLA locus mismatched related or unrelated donor HCT in a largely pediatric cohort. Graft-versus-host disease (GVHD) prophylaxis was cyclosporine with methylprednisolone. The primary end point was 1-year overall survival (OS). Thirty-four patients with HLH and 12 with other primary immune deficiencies were transplanted. With a median follow-up of 20 months, the 1-year OS for transplanted patients was 80.4% (90% confidence interval [CI], 68.6%-88.2%). Five additional deaths by 16 months yielded an 18-month OS probability of 66.7% (90% CI, 52.9%-77.3%). Two patients experienced primary graft failure, and 18 patients either experienced a secondary graft failure or required a second intervention (mostly donor lymphocyte infusion [DLI]). At 1 year, the proportion of patients alive with sustained engraftment without DLI or second HCT was 39.1% (95% CI, 25.2%-54.6%), and that of being alive and engrafted (with or without DLI) was 60.9% (95% CI, 45.4 %-74.9%). The day 100 incidence of grade II to IV acute GVHD was 17.4% (95% CI, 8.1%-29.7%), and 1-year incidence of chronic GVHD was 26.7% (95% CI, 14.6%-40.4%). Although the trial demonstrated low early mortality, the majority of surviving patients required DLI or second HCT. These results demonstrate a need for future approaches that maintain low early mortality with improved sustained engraftment. The trial was registered at Clinical Trials.gov (NCT 01998633).
- Published
- 2018
28. The Microbiome and Hematopoietic Cell Transplantation: Past, Present, and Future
- Author
-
William B. Clark, Alan Howard, Jonathan U. Peled, Daniela Weber, Pavan Reddy, John R. Wingard, Philip L. McCarthy, Marcel R.M. van den Brink, Ami S. Bhatt, Christine M. Ho, Katy Rezvani, Brahm H. Segal, Rainer Storb, Miguel-Angel Perales, John M. McCarty, Andrew Y. Koh, Takanori Teshima, Mohamad Mohty, Robert R. Jenq, Anthony D. Sung, William A. Wood, Elizabeth J. Shpall, Sophia R. Balderman, Jennifer Whangbo, Patrizia Chiusolo, Ernst Holler, Bronwen E. Shaw, Marcie L. Riches, Amin M. Alousi, Leslie S. Kean, Ryotaro Nakamura, and Tessa M. Andermann
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Prebiotic ,Hematopoietic stem cell transplantation ,Fecal microbiota transplant ,Article ,03 medical and health sciences ,medicine ,Humans ,Microbiome ,Transplantation ,Hematopoietic cell ,business.industry ,Microbiota ,Hematopoietic Stem Cell Transplantation ,Hematology ,Fecal bacteriotherapy ,Settore MED/15 - MALATTIE DEL SANGUE ,030104 developmental biology ,Metagenomics ,Immunology ,business - Published
- 2018
29. T Cell Costimulation Blockade with CTLA4-Ig (Abatacept) for Acute Gvhd Prevention in HLA Matched and Mismatched Unrelated Donor Transplantation: Results of the First Phase 2 Trial
- Author
-
Kayla Cribbin, Carol Dean, Alexandria Narayan, Kayla Betz, Aleksandra Petrovic, Maxim Norkin, Amelia Langston, Andrew C. Harris, Jeffrey H. Davis, David A. Jacobsohn, Mourad Tighiouart, Bruce R. Blazar, Leslie S. Kean, Andre Rogatko, Kyle Hebert, Michael A. Pulsipher, Catherine Bresee, Yvonne Suessmuth, Alison Yu, Sungjin Kim, Anglea Banks, Michael Grimley, C.N. Duncan, Shalini Shenoy, Kirk R. Schultz, James Rhodes, Marcelo C. Pasquini, Sung Choi, Daniel J. Hunt, Nahal R. Lalefar, Ben Watkins, Muna Qayed, Shauna Sinclair, John T. Horan, Brandi Bratrude, Scott Gillespie, Naomi Schwartz, and Roger Giller
- Subjects
Transplantation ,medicine.medical_specialty ,business.industry ,Abatacept ,Mismatched Unrelated Donor ,Hematology ,Human leukocyte antigen ,Placebo ,Gastroenterology ,T-cell costimulation ,Blockade ,Median follow-up ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
We performed a Ph2 trial in adults and children to test abatacept for AGVHD prevention (‘ABA2', Clinicaltrials # NCT01743131), based on our promising preclinical and pilot patient data. ABA2 had 2 cohorts: A) HLA-mismatched (‘7/8', n = 43), a single-arm study with pre-specified CIBMTR matched analysis (vs CNI+MTX or CNI+MTX+ATG). B) HLA-matched (‘8/8', n = 142), randomized double-blind, comparing CNI+MTX+placebo vs CNI+MTX+ABA (‘ABA'). For each ABA arm, patients received 4 doses of 10mg/kg on d -1,5,14,28. ABA2 was designed as a screening Ph2 trial, with relaxed Type 1 error (0.2) and standard Type 2 error (0.2). Power analysis assumed ABA would decrease Gr 3-4 AGVHD from 30%–>10% in 7/8s and from 20%–>10% in 8/8s. Here we report top-line results for the 7/8s (median f/u = 708d, 264-1491) and 8/8s (median f/u = 369d, 180-1175). Reduced Grade 3-4 AGVHD: ABA was associated with decreased d180 Gr 3-4 AGVHD. In 7/8s, Gr 3-4 AGVHD = 2.5% in ABA (1 event/43 patients) vs 31% (CNI+MTX) and 22% (+ATG), 1 sided p = 0.001, 0.005,). In 8/8s, Gr 3-4 AGVHD = 6.85 % (5 events/73 patients) in ABA vs 14.6% (10 events) in placebo, 1 sided p = 0.068). Reduced Grade 2-4 AGVHD in 8/8s: ABA was associated with decreased d180 Gr 2-4 AGVHD. In 7/8s, Gr 2-4 AGVHD = 42% (ABA) vs 54% (CNI+MTX) and 45% (+ATG, 1 sided p = 0.098, 0.25). In 8/8s, Gr 2-4 AGVHD = 44.5% in ABA vs 62.3% in placebo (1 sided p = 0.004). Chronic GVHD: For 7/8s, 1 yr CGVHD = 38.8% (ABA) vs 43.5% (CNI+MTX) and 35.5% (+ATG, p = 0.4, 0.99). For 8/8s, follow up is still too short to adequately evaluate, and adjudication is ongoing. No Increase In Relapse: In 7/8s, relapse = 9.37% at 1 y vs 12.9% in CNI+MTX and 13.6% in +ATG (p = 0.115 and 0.085). In 8/8s, relapse = 13.8% at 1y (ABA) vs 20.5% (placebo, p = 0.7). Remarkably, in 7/8s, where median follow up=708d, there have been no further relapses reported in ABA2, with 9.37% relapse at 2 yr vs 20.63% (CNI+MTX) and 23.4% (ATG), despite matched disease risk. Statistically significant survival advantage in 7/8s: TRM: For 7/8s, 1 yr TRM = 10.5% (ABA) vs 32.7% (CNI+MTX) and 26% (+ATG, p = 0.024, 0.365). For 8/8s, TRM = 7.1% vs 14.6% at 1 yr (p = 0.5). Relapse-Free Survival (RFS): For 7/8s, RFS = 73.7% (ABA) vs 38.7% in CNI+MTX and 48.7% in +ATG (p = 0.001 and 0.027). For 8/8s, RFS = 79.1% for ABA vs 64.9% (placebo, p = 0.38). OS: For 7/8s, OS = 71% (ABA) vs 47.5% (CNI+MTX) and 58% (+ATG, p = 0.01 and 0.145). For 8/8s, OS = 83.2% (ABA) vs 76.6 (placebo, p = 0.32). Our results suggest that short-course (4 doses) ABA can safely prevent AGVHD without compromising relapse. While this was a modestly sized study (7/8s: n = 43, 8/8 n = 142), the comparative event size for 7/8s was high enough that the protective effect of ABA was highly statistically significant. For 8/8s, there was a statistically significant improvement for Gr 2-4 GVHD and a trend toward an advantage for ABA in all other parameters. These results are the first to demonstrate efficacy of in vivo T cell costimulation blockade in preventing AGVHD.
- Published
- 2019
- Full Text
- View/download PDF
30. Immunophenotypic parameters and RBC alloimmunization in children with sickle cell disease on chronic transfusion
- Author
-
Marianne E.M. Yee, Robert Sheppard Nickel, Cassandra D. Josephson, John T. Horan, Erin K. Meyer, Ross M. Fasano, Jeanne E. Hendrickson, Leslie S. Kean, and Anne M. Winkler
- Subjects
medicine.medical_specialty ,Anemia ,business.industry ,medicine.medical_treatment ,Splenectomy ,Autoantibody ,Hematology ,medicine.disease ,Isoantibodies ,Immunophenotyping ,Internal medicine ,Immunology ,medicine ,Transfusion therapy ,Young adult ,business ,Stroke - Abstract
Alloimmunization against red blood cell (RBC) antigens is a cause of morbidity and mortality in transfused patients with sickle cell disease (SCD). To investigate distinguishing characteristics of patients who develop RBC alloantibodies after transfusion (responders) versus those who do not (non-responders), a cross-sectional study of 90 children with SCD on chronic RBC transfusion therapy at a single institution was conducted in which 18 immune parameters (including T and B cell subsets) were tested via flow cytometry, and medical records were reviewed. RBC alloimmunization was present in 26/90 (29%) patients, with anti-E, K, and C among the most commonly detected alloantibodies despite prophylactic matching for these antigens at the study institution. In addition, RBC autoantibodies had been detected in 18/26 (69%) of alloimmunized versus 7/64 (11%) of non-alloimmunized patients (P
- Published
- 2015
- Full Text
- View/download PDF
31. Use of Alefacept for Preconditioning in Multiply Transfused Pediatric Patients with Nonmalignant Diseases
- Author
-
Leslie S. Kean, Muna Qayed, Ann E. Haight, Kuang-Yueh Chiang, John T. Horan, and Elizabeth Stenger
- Subjects
Male ,Transplantation Conditioning ,medicine.medical_treatment ,Lymphocyte ,Graft vs Host Disease ,Pilot Projects ,Hematopoietic stem cell transplantation ,0302 clinical medicine ,T-Lymphocyte Subsets ,Child ,Bone Marrow Transplantation ,0303 health sciences ,Graft Survival ,Anemia, Aplastic ,Hematology ,3. Good health ,Killer Cells, Natural ,Haematopoiesis ,medicine.anatomical_structure ,Female ,Nonmalignant diseases ,Cord Blood Stem Cell Transplantation ,Stem cell ,Unrelated Donors ,medicine.drug ,Recombinant Fusion Proteins ,CD2 Antigens ,Blood Component Transfusion ,Alefacept ,Rejection ,Article ,Dyskeratosis Congenita ,Lymphocyte Depletion ,Immunophenotyping ,03 medical and health sciences ,Immune system ,medicine ,Humans ,030304 developmental biology ,Transplantation ,business.industry ,Historically Controlled Study ,Infant ,Calcineurin ,Fanconi Anemia ,Immunology ,business ,Immunologic Memory ,CD8 ,Conditioning ,030215 immunology - Abstract
Transfusion-related alloimmunization is a potent barrier to the engraftment of allogeneic hematopoietic stem cells in patients with nonmalignant diseases (NMDs). Memory T cells, which drive alloimmunization, are relatively resistant to commonly used conditioning agents. Alefacept, a recombinant leukocyte function antigen-3/IgG1 fusion protein, targets CD2 and selectively depletes memory versus naive T cells. Three multiply transfused pediatric patients with NMD received a short course of high-dose i.v. alefacept (.25 mg/kg/dose on days -40 and -9 and .5 mg/kg/dose on days -33, -26, -19, and -12) before undergoing unrelated allogeneic transplant in the setting of reduced-intensity pretransplant conditioning and calcineurin inhibitor-based post-transplant graft-versus-host disease (GVHD) prophylaxis. Alefacept infusions were well tolerated in all patients. Peripheral blood flow cytometry was performed at baseline and during and after alefacept treatment. As expected, after the 5 weekly alefacept doses, each patient demonstrated selective loss of CD2(hi)/CCR7(-)/CD45RA(-) effector memory (Tem) and CD2(hi)/CCR7(+)/CD45RA(-) central memory (Tcm) CD4(+) and CD8(+) T cells with relative preservation of the CD2(lo) Tem and Tcm subpopulations. In addition, depletion of CD2(+) natural killer (NK) cells also occurred. Neutrophil recovery was rapid, and all 3 patients had 100% sorted (CD3/CD33) peripheral blood donor chimerism by day +100. Immune reconstitution (by absolute neutrophil, monocyte, and lymphocyte counts) was comparable with a cohort of historical control patients. All 3 patients developed GVHD but are all now off immune suppression and2 years post-transplant with stable full-donor engraftment. These results suggest that alefacept at higher dosing can deplete both memory T cells and NK cells and that incorporating CD2-targeted depletion into a reduced-intensity transplant regimen is feasible and safe in heavily transfused patients.
- Published
- 2015
- Full Text
- View/download PDF
32. Immune parameter analysis of children with sickle cell disease on hydroxycarbamide or chronic transfusion therapy
- Author
-
Leslie S. Kean, John T. Horan, Daniel E. L. Promislow, David R. Archer, Jeanne E. Hendrickson, Aneesah Garrett, Ifeyinwa Osunkwo, Robert Sheppard Nickel, and Jennifer Robertson
- Subjects
Blood transfusion ,Adolescent ,medicine.medical_treatment ,Cell ,Anemia, Sickle Cell ,Disease ,Article ,Hydroxycarbamide ,Leukocyte Count ,Immune system ,Immunophenotyping ,Antisickling Agents ,Leukocytes ,medicine ,Humans ,Hydroxyurea ,Blood Transfusion ,Child ,Immune phenotype ,business.industry ,Hematology ,Black or African American ,medicine.anatomical_structure ,Immunology ,Transfusion therapy ,business ,Follow-Up Studies ,medicine.drug - Abstract
Sickle cell disease (SCD) is increasingly appreciated as an inflammatory condition associated with alterations in immune phenotype and function. In this cross-sectional study we performed a multiparameter analysis of 18 immune markers in 114 paediatric SCD patients divided by treatment group [those receiving hydroxycrabamide (HC, previously termed hydroxyurea), chronic transfusion (CT), or no disease-modifying therapy] and 29 age-matched African American healthy controls. We found global elevation of most immune cell counts in SCD patients receiving no disease-modifying therapy at steady state. Despite the decrease in percentage of haemoglobin S associated with CT therapy, the abnormal cellular immune phenotype persisted in patients on CT. In contrast, in both univariate and multivariate analysis, treatment with HC was associated with normalization of the vast majority of leucocyte populations. This study provides additional support for HC treatment in SCD, as it appears that HC decreases the abnormally elevated immune cell counts in patients with SCD.
- Published
- 2015
- Full Text
- View/download PDF
33. Successful treatment of pediatric CML in Cambodia: an international collaboration
- Author
-
Scott C. Howard, Sreng Kea, Sing Heng, Ibrahim Qaddoumi, Rathi Guhadasan, Leslie S. Kean, Prom Vireak, Hour Putchat, Katherine Tarlock, Bruce M. Camitta, Benjamin Watkins, and Sath Sainghan
- Subjects
medicine.medical_specialty ,business.industry ,Perinatal complications ,medicine ,Developing country ,Cancer ,Hematology ,Intensive care medicine ,business ,medicine.disease ,Pediatric cancer ,Global Capacity-Building Showcase - Abstract
Treating children with cancer is challenging in developing countries such as Cambodia. The large burden of mortality and morbidity from infectious diseases and perinatal complications leaves few resources for pediatric cancer programs. Angkor Hospital for Children (AHC) is a Cambodian pediatric
- Published
- 2017
34. Chimeric Antigen Receptor T Cell-Mediated Neurotoxicity in Nonhuman Primates
- Author
-
Daniel J. Hunt, Bruce R. Blazar, Hannah Brakke, Luis F. Gonzalez-Cuyar, Rebecca Gardner, Olivia Finney, Scott N. Furlan, H. Denny Liggitt, Rafael Ponce, Agne Taraseviciute, Stanley R. Riddell, Audrey Baldessari, Alison Yu, Carolina Berger, David Myerson, Virginia Hoglund, Jessica M. Snyder, Hengqi Zheng, Michael C. Jensen, Chris English, Leslie S. Kean, Victor Tkachev, and Cameron J. Turtle
- Subjects
0301 basic medicine ,Adoptive cell transfer ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell ,Immunotherapy, Adoptive ,Transplantation, Autologous ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cyclophosphamide ,CD20 ,biology ,business.industry ,Neurotoxicity ,Immunotherapy ,medicine.disease ,Antigens, CD20 ,Macaca mulatta ,Chimeric antigen receptor ,Cytokine release syndrome ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Neurotoxicity Syndromes ,business ,K562 Cells - Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy has revolutionized the treatment of refractory leukemias and lymphomas, but is associated with significant toxicities, namely cytokine release syndrome (CRS) and neurotoxicity. A major barrier to developing therapeutics to prevent CAR T cell–mediated neurotoxicity is the lack of clinically relevant models. Accordingly, we developed a rhesus macaque (RM) model of neurotoxicity via adoptive transfer of autologous CD20-specific CAR T cells. Following cyclophosphamide lymphodepletion, CD20 CAR T cells expand to 272 to 4,450 cells/μL after 7 to 8 days and elicit CRS and neurotoxicity. Toxicities are associated with elevated serum IL6, IL8, IL1RA, MIG, and I-TAC levels, and disproportionately high cerebrospinal fluid (CSF) IL6, IL2, GM-CSF, and VEGF levels. During neurotoxicity, both CD20 CAR and non-CAR T cells accumulate in the CSF and in the brain parenchyma. This RM model demonstrates that CAR T cell–mediated neurotoxicity is associated with proinflammatory CSF cytokines and a pan–T cell encephalitis. Significance: We provide the first immunologically relevant, nonhuman primate model of B cell–directed CAR T-cell therapy–mediated CRS and neurotoxicity. We demonstrate CAR and non-CAR T-cell infiltration in the CSF and in the brain during neurotoxicity resulting in pan-encephalitis, accompanied by increased levels of proinflammatory cytokines in the CSF. Cancer Discov; 8(6); 750–63. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 663
- Published
- 2017
35. Future of allogeneic hematopoietic stem cell transplantation for chemotherapy-resistant pediatric acute leukemia: potential advances
- Author
-
Leslie S. Kean and Marie Bleakley
- Subjects
Oncology ,Chemotherapy ,medicine.medical_specialty ,Acute leukemia ,business.industry ,medicine.medical_treatment ,Myeloid leukemia ,Hematology ,Hematopoietic stem cell transplantation ,Donor Lymphocytes ,medicine.disease ,Transplantation ,Leukemia ,surgical procedures, operative ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Pharmacology (medical) ,business ,Chemoradiotherapy - Abstract
Fred Hutchinson Cancer Research Center, Seattle, WA, USA University of Washington Department of Pediatrics, Seattle, WA, USA Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, USA *Author for correspondence: Tel.: +1 206 667 6572; Fax: +1 206 667 7983; mbleakle@fhcrc.org Transplantation as a curative strategy for high-risk leukemia Allogeneic hematopoietic stem cell transplantation (HCT) is curative for many children with life-threatening, chemotherapy-resistant leukemia. HCT works by two major mechanisms: high-intensity chemotherapy, or chemoradiotherapy pretransplant conditioning; and an immunologic antileukemia effect mediated by donor lymphocytes in the graft, the ‘graftversus-leukemia effect’ (GVL). Survival rates for pediatric leukemia HCT recipients have steadily increased over time with current overall survival rates for pediatric acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) of >50%. The improvements in survival principally reflect major reductions in early postHCT treatment-related mortality (TRM) that have occurred as a result of better supportive care. The decrease in TRM in unrelated donor, cord blood (CBT) and mismatched family donor HCT has been particularly notable, and survival rates after ‘alternative donor HCT’ are now broadly comparable to HLA-matched sibling donor HCT. Unfortunately, relatively little reduction has been achieved in relapse rates, making relapse now the most frequent cause of failure of HCT for leukemia [1]. Relapse occurs in 10–30% of HCTs performed for pediatric leukemia in remission and has a dismal prognosis: the median survival of children with postHCT relapse is 4 months [2]. In order to further improve overall survival after allogeneic HCT for pediatric leukemia, novel strategies to address post-HCT leukemia relapse are crucial.
- Published
- 2014
- Full Text
- View/download PDF
36. Transcriptomic Analysis of CD4+ T Cell Dysfunction during Gvhd: Evidence for Profound Reprograming of T Cell Signaling during Acute Gvhd That Is Controlled during CD28:CD80/86 Costimulation Blockade with Abatacept
- Author
-
Kayla Betz, Leslie S. Kean, Alison Yu, John T. Horan, Kathryn L. Pellegrini, Amelia Langston, Muna Qayed, Steven E. Bosinger, Yvonne Suessmuth, Benjamin Watkins, Bruce R. Blazar, and Brandi Bratrude
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Abatacept ,T cell ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,Calcineurin ,medicine.anatomical_structure ,Graft-versus-host disease ,Internal medicine ,Lymphocyte costimulation ,Medicine ,business ,CD80 ,medicine.drug - Abstract
Although acute graft-versus-host-disease (AGVHD) is one of the major causes of non-relapse mortality after hematopoietic stem cell transplant (HCT), we are still unable to predict which patients will develop the most severe form of this disease, or which molecular pathways are dysregulated in the T cells that cause disease. Thus, understanding the molecular features of AGVHD is a critical unmet need. To address this, we have performed a companion mechanistic study as a part of our completed Phase 2 trial of abatacept, a CD28:CD80/86 costimulation blockade agent, for severe AGVHD prevention (Clinicaltrials.gov # NCT01743131, 'ABA2'). ABA2 has demonstrated significant improvement in AGVHD in patients prophylaxed with abatacept in addition to calcineurin inhibition (CNI) + Methotrexate (MTX) compared to controls receiving CNI/MTX alone. To begin to uncover mechanisms responsible for the control of AGVHD with abatacept, and given that CD4+ T cells have been consistently documented to be the main therapeutic target of this drug, we interrogated the transcriptome of CD4+ T cells reconstituting in patients prophylaxed with abatacept compared to CNI/MTX. To perform this analysis, we flow cytometrically sorted CD4+ T cells on Days 21-28 post-transplant from all patients on ABA2, as well as a cohort of 12 untransplanted healthy controls, and subsequently performed mRNA-sequencing on these cells. Weighted Gene Correlation Network Analysis (WGCNA) was performed on the top 6000 most variant transcripts from the resulting sequencing data. Hierarchical clustering of the WGCNA co-expression matrix enabled the identification of self-assembling modules (SAMs) that met a threshold of coexpression (Figure 1A). For the ABA2 dataset, we considered the following variables in the WGCNA model: patient cohort (7/8 patients, 8/8 patients, healthy controls), +/- prophylaxis with abatacept, CMV reactivation, EBV reactivation, Grade of GVHD (0-4), relapse, non-relapse mortality, and all-cause mortality. The WGCNA clustering analysis resulted in the identification of 4 discrete SAMs, which were highly correlated with clinical variable metamodules. This analysis revealed a strong positive correlation of a 476-gene SAM (the Turquoise module) in patients prophylaxed with CNI/MTX + placebo and anti-correlation of this module in patients prophylaxed with CNI/MTX + abatacept, as demonstrated in both the WGCNA heatmap and through Gene Set Enrichment Analysis (Figure 1 A-B). These opposing correlations suggested that interrogation of this module would reveal mechanistic correlates with standard prophylaxis that were decoupled by abatacept. Pathway analysis using the Reactome database (Figure 1C) revealed the turquoise SAM to be dominated by four types of pathways: (1) Those that define canonical cell-cycle pathways (2) Those involved in T cell metabolism (3) Those involved in apoptosis and (4) Those involved in T cell activation, consistent with upregulation of these transcripts in placebo versus abatacept patients. In addition to being highly correlated with patients receiving placebo, the expression of a subset of the transcripts in the Turquoise module were also directly correlated with the severity of AGVHD in these patients. Thus, linear regression analysis of the 476 transcripts in this module identified a subset of 93 genes for which transcript expression level was increased both in placebo compared to abatacept, and for which expression level also positively correlated with Grade of AGVHD. As with the Turquoise module as a whole, this subset of genes also formed a highly correlated network, linking transcripts involved in T cell proliferation, apoptosis, activation, metabolism as well as the T cell checkpoint (Figure 1D). This analysis represents the first comprehensive interrogation of the transcriptomic correlates of AGVHD. It identifies a novel set of transcripts which positively associate with the severity of AGVHD, and which costimulation blockade with abatacept down-regulates and de-couples from AGVHD severity. These results suggest a profound reprograming of T cell activation with abatacept that is correlated with the control of AGVHD. Disclosures Qayed: Bristol-Myers Squibb: Honoraria. Langston:Astellas Pharma: Other: Research Support; Incyte: Other: Research Support; Jazz Pharmaceuticals: Other: Research Support; Chimerix: Other: Research Support; Takeda: Other: Research Support; Kadmon Corporation: Other: Research Support; Novartis: Other: Research Support; Bristol Myers Squibb: Other: Research Support. Blazar:Fate Therapeutics, Inc.: Research Funding; RXi Pharmaceuticals: Research Funding; Alpine Immune Sciences, Inc.: Research Funding; Abbvie Inc: Research Funding; Leukemia and Lymphoma Society: Research Funding; Childrens' Cancer Research Fund: Research Funding; KidsFirst Fund: Research Funding; Tmunity: Other: Co-Founder; BlueRock Therapeutics: Membership on an entity's Board of Directors or advisory committees; Kamon Pharmaceuticals, Inc: Membership on an entity's Board of Directors or advisory committees; Five Prime Therapeutics Inc: Co-Founder, Membership on an entity's Board of Directors or advisory committees; Regeneron Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Magenta Therapeutics and BlueRock Therapeuetics: Membership on an entity's Board of Directors or advisory committees. Kean:HiFiBio: Consultancy; BlueBirdBio: Research Funding; Gilead: Research Funding; Regeneron: Research Funding; EMDSerono: Consultancy; FortySeven: Consultancy; Magenta: Research Funding; Kymab: Consultancy; Jazz: Research Funding; Bristol Meyers Squibb: Patents & Royalties, Research Funding. OffLabel Disclosure: Abatacept: Approved for Rheumatoid Arthritis; used in this trial for prevention of GVHD.
- Published
- 2019
- Full Text
- View/download PDF
37. Making Progress in Graft-Versus-Host Disease Prophylaxis and Microbiome Analysis in the Blood and Marrow Transplant Clinical Trials Network: Progress III (1703)/MI-Immune (1801)
- Author
-
Miguel-Angel Perales, Wael Saber, Javier Bolaños-Meade, Ami S. Bhatt, Shernan G. Holtan, Leslie S. Kean, Mehdi Hamadani, Brent R. Logan, and Raphael Fraser
- Subjects
business.industry ,Surrogate endpoint ,Immunology ,Human microbiome ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Tacrolimus ,Clinical trial ,Transplantation ,Graft-versus-host disease ,Immune system ,Medicine ,Microbiome ,business ,health care economics and organizations - Abstract
Graft-versus-host disease (GVHD) remains the major source of morbidity and transplant-related mortality after allogeneic hematopoietic cell transplant (alloHCT). Since the mid-1980s, the standard prophylaxis for GVHD has been a calcineurin inhibitor plus methotrexate (MTX). Subsequent clinical trials designed to intensify GVHD prophylaxis have shown an increased risk of relapse, thus negating survival benefit. The ideal GVHD prophylaxis regimen is therefore associated with a high graft-versus-host disease-free, relapse-free survival (GRFS), a composite endpoint that reflects survival without morbidity. A recent 3-arm randomized phase II study conducted through the Blood and Marrow Transplant Clinical Trials Network (BMT CTN 1203) showed a benefit in GRFS using post-transplant cyclophosphamide, tacrolimus, and mycophenolate mofetil (PTCy/Tac/MMF) with a hazard ratio of 0.72 (90% confidence interval 0.54 - 0.94, p-0.044) compared to contemporary non-randomized CIBMTR Tac/MTX controls. Based upon these results, the BMT CTN has launched 1703, a multicenter randomized phase III study of standard Tac/MTX vs PTCy/Tac/MMF in the setting of reduced intensity conditioning, matched peripheral blood stem cell transplantation (RIC PBSCT). The primary endpoint of 1703 is GRFS at 1 year, and several secondary endpoints, including patient-reported outcomes, will be analyzed. A series of recent, predominantly single-center studies have shown an association with the composition of intestinal microbiota and the development of acute GVHD, which suggests that this may be a potential modifiable biomarker of disease. The generalizability of findings from these studies is yet unknown, and no large multi-center studies on the intestinal microbiota of HCT patients has yet been carried out. Patients enrolled in 1703 will be eligible to co-enroll in BMT CTN 1801, the Mi-Immune study, detailing microbiota and immune reconstitution in this cohort. The primary endpoint of 1801 is 1-year non-relapse mortality by tertiles of stool microbial diversity at engraftment. Secondary objectives, including effect of GVHD prophylaxis on diversity, oligodomination and risk of bloodstream infection, volume of antimicrobial exposure, and T-cell receptor diversity, will be analyzed. The combined studies of BMT CTN 1703/1801 will define the standard of care for GVHD prophylaxis in the RIC PBSCT setting and vastly expand our knowledge on the impact of intestinal microbiota in critical outcomes after alloHCT. This clinical trial is registered at ClinicalTrials.gov NCT03959241. Figure Disclosures Holtan: Incyte: Consultancy; Bristol-Myers Squibb: Consultancy; Janssen: Consultancy; CSL Behring: Consultancy. Bhatt:ArcBio: Other: Scientific Advisory Board; January.ai: Other: Scientific Advisory Board; Caribou Bioscience: Other: Scientific Advisory Board; Kaleido Biosciences: Consultancy, Other: Paid Consultant; Janssen Human Microbiome Institute: Consultancy, Other: Paid Consultant; Illumina: Honoraria; 10x Genomics: Other: Research collaboration without funding support; Illumina: Other: Research collaboration without funding support; Agilent: Research Funding; Global Oncology, Inc: Other: Nonprofit Board. Perales:Bellicum: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol-Meyers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; MolMed: Membership on an entity's Board of Directors or advisory committees; NexImmune: Membership on an entity's Board of Directors or advisory committees; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees; Omeros: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Consultancy, Honoraria; Medigene: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees; Kyte/Gilead: Research Funding; Miltenyi: Research Funding; Incyte: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Nektar Therapeutics: Honoraria, Membership on an entity's Board of Directors or advisory committees. Kean:HiFiBio: Consultancy; BlueBirdBio: Research Funding; Gilead: Research Funding; Regeneron: Research Funding; EMDSerono: Consultancy; FortySeven: Consultancy; Magenta: Research Funding; Kymab: Consultancy; Jazz: Research Funding; Bristol Meyers Squibb: Patents & Royalties, Research Funding. Hamadani:Medimmune: Consultancy, Research Funding; Takeda: Research Funding; Pharmacyclics: Consultancy; Janssen: Consultancy; Merck: Research Funding; Celgene: Consultancy; Otsuka: Research Funding; ADC Therapeutics: Consultancy, Research Funding; Sanofi Genzyme: Research Funding, Speakers Bureau. Bolaños-Meade:Incyte Corporation: Other: DSMB fees.
- Published
- 2019
- Full Text
- View/download PDF
38. An All Antibody Approach for Conditioning Bone Marrow for Hematopoietic Stem Cell Transplantation with Anti-cKIT and Anti-CD47 in Non-Human Primates
- Author
-
Jie Liu, Sharline Chen, Cynthia E. Dunbar, Irving L. Weissman, James Y. Chen, Balaji Agoram, Ravi Majeti, Kristopher D. Marjon, Chris H. Takimoto, Jiaqi Duan, Kavitha Sompalli, Timothy S Choi, Mark P. Chao, Mwe Mwe Chao, Jens-Peter Volkmer, Leslie S. Kean, Dongdong Feng, and Ofelia Mata
- Subjects
biology ,medicine.drug_class ,business.industry ,Genetic enhancement ,medicine.medical_treatment ,CD47 ,Immunology ,Stem cell factor ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Monoclonal antibody ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,medicine ,biology.protein ,Cancer research ,Bone marrow ,Antibody ,business - Abstract
Background Hematopoietic stem cell (HSC) transplantation (HSCT) is a well-established procedure that, with or without gene therapy, is curative for numerous severe life-threatening diseases including genetic blood disorders and blood cancers. While advances have been made, there are still substantial concerns since these chemo- and radiation therapy based procedures cause long-term toxicities such as infertility and secondary malignancies or even result in high mortality. We have previously established in a series of preclinical studies a novel chemo- and radiation-free non-toxic monoclonal antibody (Ab) -based conditioning regimen for autologous and allogeneic HSCT (Czechowicz et al., Akanksha et al. and George et al.). This cKIT-CD47 Ab-based regimen selectively depletes host HSCs for HSCT while sparing off-target toxicities caused by chemotherapy/radiation. By significantly decreasing morbidity/mortality associated with traditional conditioning regimens, antibody-mediated conditioning could expand the patient population eligible to receive HSCT for a variety of disorders. We developed a novel cKIT Ab (FSI-174), with an active Fc, and in combination with our CD47 magrolimab (previously 5F9, blocks the don't eat me pathway) could be utilized to translate the promising preclinical findings into clinical studies for safe and less toxic bone marrow conditioning for HSCT. Here we present the functional characterization of FSI-174 as single Ab and in combination with magrolimab in vitro and in non-human primate (NHP) studies. Methods We tested if FSI-174 could block stem cell factor signaling and we explored if FSI-174 alone or in combination with magrolimab could promote phagocytosis of cKIT positive cells (Kasumi-1). In addition, we determined if FSI-174 could cause mast cell degranulation. Subsequently, we explored the potential of FSI-174 alone (Phase A) or in combination with magrolimab (Phase B) to deplete HSCs in NHPs (rhesus macaques)in vivo. In Phase A, single doses of FSI-174 (0.3, 1, or 3 mg/kg) were administered alone. In Phase B, FSI-174 (0.3 or 3 mg/kg) was administered in combination with magrolimab (5mg/kg priming and 20 mg/kg maintenance dose). Bone marrow aspirates and core biopsies and peripheral blood were sampled before the study start and throughout the study. Frequency of bone marrow HSCs and cKIT receptor occupancy (RO) was determined by flow cytometry. In addition, the PK profile of FSI-174 was determined. Results In-vitro analysis demonstrated that FSI-174 decreases proliferation of HSPCs and enhances phagocytosis of cKIT positive cells, and the addition of magrolimab synergistically enhances the phagocytosis. Strikingly, FSI-174 did not cause mast cell degranulation in vitro. In the NHPs, complete (100%) cKIT receptor occupancy was achieved at all FSI-174 dose levels and was maintained for 1 to 9 days correlating with increasing doses and pharmacokinetics. The FSI-174 Cmax was found to be proportional to dose and mean Cmax increased from 6.25 ug/mL to 49.2 ug/mL. In Phase A, FSI-174 alone did not decrease the frequency of bone marrow HSCs compared to PBS control and had no effect on the peripheral blood cell counts. However, in Phase B, when FSI-174 was combined with magrolimab it significantly decreased the frequency of bone marrow HSCs with the nadir at day 9 and no recovery over 85 days compared to PBS control. Notably, there were no changes in peripheral blood cell counts over the course of the studies with no cytopenias in combination treatment. Conclusions We have developed a novel cKIT Ab (FSI-174) that meets the desired profile of stem cell factor block, promotion of phagocytosis, but without promoting mast cell degranulation. Furthermore, in the NHPs studies we have confirmed our chemo- and radiation-free cKIT-CD47 Ab -based conditioning approach with FSI-174 and magrolimab. As anticipated by our previous preclinical studies, monotherapy with FSI-174 does not deplete bone marrow HSCs in NHPs. Notably, no cytopenias are observed with either monotherapy or combination therapy. These data demonstrate the specificity, efficacy and safety of FSI-174/ magrolimab combination have great potential for conditioning regimen for HSCT in a chemotherapy and radiation free manner. Given the favorable safety profile of magrolimab across several clinical studies, these results are paving the way to the first-in-human trials for this novel conditioning for HSCT. Disclosures Marjon: Forty Seven Inc: Employment, Equity Ownership. Chen:Forty Seven Inc.: Consultancy, Equity Ownership. Duan:Forty Seven Inc.: Employment, Equity Ownership. Choi:Forty Seven inc: Employment, Equity Ownership. Sompalli:Forty Seven Inc: Employment, Equity Ownership. Feng:Forty Seven Inc: Employment, Equity Ownership. Mata:Forty Seven inc: Employment, Equity Ownership. Chen:Forty Seven Inc: Employment, Equity Ownership. Kean:HiFiBio: Consultancy; BlueBirdBio: Research Funding; Gilead: Research Funding; Regeneron: Research Funding; EMDSerono: Consultancy; FortySeven: Consultancy; Magenta: Research Funding; Bristol Meyers Squibb: Patents & Royalties, Research Funding; Kymab: Consultancy; Jazz: Research Funding. Chao:Forty Seven Inc: Employment, Equity Ownership. Chao:Forty Seven, Inc.: Employment, Equity Ownership, Patents & Royalties. Takimoto:Forty Seven, Inc.: Employment, Equity Ownership, Patents & Royalties. Agoram:Forty Seven Inc.: Employment, Equity Ownership. Majeti:FortySeven: Consultancy, Equity Ownership, Other: Board of Director; BioMarin: Consultancy. Weissman:Forty Seven Inc.: Consultancy, Equity Ownership, Patents & Royalties. Liu:Forty Seven Inc: Employment, Equity Ownership, Patents & Royalties. Volkmer:Forty Seven, Inc.: Employment, Equity Ownership, Patents & Royalties.
- Published
- 2019
- Full Text
- View/download PDF
39. Equate, a Phase 1b/2 Study Evaluating the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Clinical Activity of a Novel Targeted Anti-CD6 Therapy, Itolizumab, in Subjects with Newly Diagnosed Acute Graft Versus Host Disease
- Author
-
Madan Jagasia, Saurabh Chhabra, Mark Sonnemann, Thomas C. Shea, Stephen Connelly, Daanish Hoda, Leslie S. Kean, Corey Cutler, George Chen, Susan Light, Cherie Tracy Ng, Gabrielle Meyers, Joseph Pidala, John Koreth, Betty K. Hamilton, Jerome Ritz, Robert J. Soiffer, Ryotaro Nakamura, Krishna Polu, Edmund K. Waller, David Essayan, Nuwan Nanayakkara, Marco Mielcarek, and Joel Rothman
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Surrogate endpoint ,business.industry ,medicine.medical_treatment ,Immunology ,Itolizumab ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Placebo ,medicine.disease ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Graft-versus-host disease ,Tolerability ,Internal medicine ,medicine ,Clinical endpoint ,Cumulative incidence ,business ,030215 immunology ,medicine.drug - Abstract
Background: Acute graft versus host disease (aGVHD) occurs in approximately 50% of patients receiving allogeneic hematopoietic stem cell transplant (HSCT) with standard GVHD prophylaxis regimens and is a significant contributor to morbidity and non-relapse mortality (NRM). There are currently no FDA approved therapies for the initial treatment of newly diagnosed aGVHD. T effector cells (Teff), particularly CD4+ T helper cells, play a central role in the pathogenesis of aGVHD. CD6 is a co-stimulatory receptor, predominantly expressed on CD4+ T helper cells, that binds to activated leukocyte cell adhesion molecule (ALCAM), a ligand expressed on antigen presenting cells and various epithelial and endothelial tissues. The CD6-ALCAM pathway plays an integral role in modulating T cell activation and trafficking and is central to immune mediated inflammation. Previous studies by Soiffer et al. in patients receiving allogeneic bone marrow transplants had shown that depleting CD6+ T cells ex-vivo in donor bone marrow using T-12, an anti-CD6 monoclonal antibody (mAb), as a sole method for GVHD prophylaxis lowered the incidence of acute and chronic GVHD. EQ001 (itolizumab) is a humanized IgG1 mAb that binds to CD6 and blocks the interaction with ALCAM and inhibits both the activity and trafficking of T cells without causing T cell depletion. Itolizumab has s previously been developed and is approved for the treatment of Psoriasis in India. The potential effectiveness of an anti-CD6 mAb targeted approach in treating aGVHD is supported by data from pre-clinical models evaluating the prevention and treatment of GVHD with EQ001. The objective of this study is to evaluate the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and clinical activity of EQ001 in subjects with newly diagnosed aGVHD. Study Design and Methods: This is a multi-center Phase 1b/2 study (NCT03763318) of EQ001 in subjects with aGVHD. Part A (Phase 1b) of the study is an open-label, multiple ascending dose study of EQ001 in adults with newly diagnosed High Risk (MacMillan Criteria) aGVHD following HSCT. Approximately 24 subjects will be enrolled in successive cohorts of 3 to 6 adult subjects using a standard 3 + 3 dose escalation design. For each cohort, subjects will receive EQ001 administered intravenously (IV) every two weeks for five doses with corticosteroids (CS) tapered over the treatment period. A total of four dose levels will be tested ranging from 0.4 mg/kg to 2.4 mg/kg, and subjects will be followed for an additional 10 months after study treatment is completed. The primary objective of Part A is to evaluate safety and tolerability of EQ001. Secondary objectives include an evaluation of PK, PD (including CD6 receptor occupancy, changes in inflammatory cytokines and novel gut biomarkers [ST2, REG3α], and changes in Teff/T regulatory cells populations), and clinical activity (overall aGVHD response rates on Day 29, durability of the response at Day 57 and up to 1 year, cumulative incidence of NRM, rates of chronic GVHD, and CS dose requirements). Data from Part A will inform the dose selection for Part B (Phase 2) of the study. Enrollment to Part A is ongoing. Part B is a double-blind, placebo-controlled study in subjects (age ≥12) with newly diagnosed Grade II-IV aGVHD. Approximately 60 subjects will be randomized in a 2:1 ratio to either treatment with EQ001 (n=40) or placebo (n=20) in addition to and within 72 hours of the first dose of CS. EQ001 or placebo will be administered IV every two weeks for five doses, and subjects will be followed for an additional 10 months after the last dose of study treatment. The primary objective of Part B is to evaluate the overall response rate of aGVHD in patients treated with EQ001 versus placebo. Secondary objectives include an evaluation of safety and other clinical endpoints. The primary endpoint is the overall response rate at Day 29 as defined by a complete response, very good partial response or partial response. Additional clinical endpoints include assessment of NRM, chronic GVHD rates, and CS dose requirements. Conclusion: Inhibiting the CD6-ALCAM pathway with EQ001 represents a unique and promising approach for the treatment of aGVHD following HSCT by inhibiting both the activity and trafficking of T cells. This is the first clinical study to examine the effects of an anti-CD6 therapy for the treatment of patients with newly diagnosed aGVHD. Disclosures Koreth: Equillium: Consultancy; Cugene: Consultancy; Amgen: Consultancy. Shea:Jazz: Consultancy; Amgen: Consultancy; Merck: Consultancy. Jagasia:Kadmon: Consultancy; Incyte: Consultancy; Janssen: Research Funding. Waller:Pharmacyclics: Other: Travel expenses, Research Funding; Cerus Corporation: Other: Stock, Patents & Royalties; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Chimerix: Other: Stock; Cambium Oncology: Patents & Royalties: Patents, royalties or other intellectual property ; Amgen: Consultancy; Kalytera: Consultancy. Nakamura:Celgene: Other: support for an academic seminar in a university in Japan; Merck: Membership on an entity's Board of Directors or advisory committees; Alexion: Other: support to a lecture at a Japan Society of Transfusion/Cellular Therapy meeting ; Kirin Kyowa: Other: support for an academic seminar in a university in Japan. Ng:Equillium: Employment, Equity Ownership. Sonnemann:Equillium Inc.: Employment. Light:Equillium Inc.: Consultancy. Nanayakkara:Equillium Inc.: Consultancy. Essayan:Equillium Inc.: Consultancy. Rothman:Equillium Inc.: Employment, Equity Ownership. Connelly:Equillium: Employment, Equity Ownership. Polu:Equillium Inc.: Employment, Equity Ownership. Kean:HiFiBio: Consultancy; BlueBirdBio: Research Funding; Gilead: Research Funding; Regeneron: Research Funding; EMDSerono: Consultancy; FortySeven: Consultancy; Magenta: Research Funding; Kymab: Consultancy; Jazz: Research Funding; Bristol Meyers Squibb: Patents & Royalties, Research Funding. Ritz:Draper Labs: Consultancy; Talaris Therapeutics: Consultancy; TScan Therapeutics: Consultancy; Equillium: Research Funding; Merck: Research Funding; Kite Pharma: Research Funding; Aleta Biotherapeutics: Consultancy; Celgene: Consultancy; Avrobio: Consultancy; LifeVault Bio: Consultancy. Soiffer:Gilead, Mana therapeutic, Cugene, Jazz: Consultancy; Juno, kiadis: Membership on an entity's Board of Directors or advisory committees, Other: DSMB; Kiadis: Other: supervisory board; Mana therapeutic: Consultancy; Cugene: Consultancy; Jazz: Consultancy. Cutler:Kadmon: Consultancy; Omeros: Consultancy; ElsaLys: Consultancy; BiolineRx: Other: DSMB; Cellect: Other: DSMB; Kalytera: Other: DSMB; Incyte: Consultancy; Genentech: Consultancy; Jazz: Consultancy; BMS: Consultancy; Fate Therapeutics: Consultancy; Pharmacyclics: Consultancy. OffLabel Disclosure: EQ001 (itolizumab) is an investigational agent being developed for the treatment of acute GVHD. Itolizumab has not received regulatory approval for the treatment of GVHD. The abstract being submitted describes the study design for the phase 1b/2 study. No data is being presented.
- Published
- 2019
- Full Text
- View/download PDF
40. P154 SINGLE-CELL ANALYSIS OF T CELL PATHOGENESIS IN PEDIATRIC CROHN’S DISEASE
- Author
-
Leslie S. Kean, Brandi Bratrude, David L. Suskind, Carol Dean, Benjamin Doran, Jose Ordovas-Montanes, Alex K. Shalek, Victor Tkachev, Hengqi Zheng, Lucrezia Colonna, Ghassan Wahbeh, Kayla Cribbin, Scott N. Furlan, Dale Lee, Alison Yu, Lusine Ambartsumyan, and Kayla Betz
- Subjects
Pathogenesis ,medicine.anatomical_structure ,Hepatology ,Single-cell analysis ,Pediatric Crohn's disease ,business.industry ,T cell ,Immunology ,medicine ,Gastroenterology ,Immunology and Allergy ,business - Published
- 2019
- Full Text
- View/download PDF
41. Characterization of natural killer cells expressing markers associated with maturity and cytotoxicity in children and young adults with sickle cell disease
- Author
-
Nurah Lawal, Robert Sheppard Nickel, Leslie S. Kean, Marcus Dean, Emily Riehm Meier, Allistair Abraham, Yunfei Wang, Barbara Speller-Brown, Haili Lang, and Catherine M. Bollard
- Subjects
Adult ,Cytotoxicity, Immunologic ,Male ,Adolescent ,Cell ,Anemia, Sickle Cell ,Article ,Immunophenotyping ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Antisickling Agents ,hemic and lymphatic diseases ,medicine ,Humans ,Hydroxyurea ,Child ,Cytotoxicity ,Receptor ,business.industry ,Degranulation ,Infant ,Hematology ,Prognosis ,NKG2D ,Killer Cells, Natural ,Red blood cell ,Cross-Sectional Studies ,Cell killing ,medicine.anatomical_structure ,Oncology ,Case-Control Studies ,Child, Preschool ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Immunology ,Female ,business ,Biomarkers ,Follow-Up Studies ,030215 immunology - Abstract
Background Sickle cell disease (SCD) is increasingly recognized as a red blood cell disorder modulated by abnormally increased inflammation. We have previously shown that in patients with SCD not on a disease-modifying therapy (hydroxyurea or chronic transfusions), natural killer (NK) cell numbers are increased. In the current study, we further investigated the NK cell function to determine if there was evidence of increased activation and cytotoxicity. Procedure We conducted a cross-sectional study of 44 patients with HbSS/HbSβ0 thalassemia at steady state (hydroxyurea = 13, chronic transfusion = 11, no disease-modifying therapy = 20) and 23 healthy controls. Using a fresh blood sample, NK immunophenotyping was performed as follows: NK cells (CD3- CD56+ lymphocytes) were evaluated for makers associated with activation (NKG2D, NKp30, NKp44, and CD69) and maturity (CD57, killer immunoglobulin-like receptors (KIR), and CD56dim). Degranulation and cytotoxicity assays were performed to evaluate NK cell function. Results Patients with SCD who were not on disease-modifying therapy had a higher number of NK cells with an immunophenotype associated with increased cytotoxicity (NKG2D+ , NKp30+ , CD56dim+ , and KIR+ NK cells) compared with healthy controls and patients on hydroxyurea. NK cells from SCD patients not on disease-modifying therapy demonstrated significantly increased cytotoxicity (measured by assaying NK cell killing of the K562 cell line) compared with healthy controls (P = 0.005). Notably, NK cell cytotoxicity against K562 cells in the hydroxyurea or chronic transfusion patients was not significantly different from that in healthy controls. Conclusion SCD is associated with increased NK cell function as well as increased NK cell numbers, which appears to be normalized with disease-modifying therapy.
- Published
- 2019
- Full Text
- View/download PDF
42. In Vivo T Cell Costimulation Blockade with Abatacept for Acute Graft-versus-Host Disease Prevention: A First-in-Disease Trial
- Author
-
Jennifer Robertson, Amelia Langston, Divya Koura, John T. Horan, Muna Qayed, Jennifer Carr, J. Cheeseman, Aneesh K. Mehta, Leslie S. Kean, Yvonne Suessmuth, Christian P. Larsen, Heather R. Johnson, Jason A. Conger, Edmund K. Waller, Aneesah Garrett, R. Donald Harvey, Linda Stempora, Allan D. Kirk, Hanna Jean Khoury, Cynthia Couture, Audrey Grizzle, and Brandi E. Johnson
- Subjects
Adult ,Male ,musculoskeletal diseases ,medicine.medical_specialty ,Immunoconjugates ,Transplantation Conditioning ,Adolescent ,T-Lymphocytes ,T cell ,Graft vs Host Disease ,Gastroenterology ,Abatacept ,Young Adult ,Costimulation blockade ,Internal medicine ,Humans ,Transplantation, Homologous ,Medicine ,Aged ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,Graft-versus-host disease prophylaxis ,Middle Aged ,Blockade ,surgical procedures, operative ,medicine.anatomical_structure ,Allogeneic transplantation ,Pharmacodynamics ,Acute Disease ,Cohort ,Immunology ,Feasibility Studies ,Female ,Methotrexate ,business ,Immunosuppressive Agents ,CD80 ,medicine.drug - Abstract
We performed a first-in-disease trial of in vivo CD28:CD80/86 costimulation blockade with abatacept for acute graft-versus-host disease (aGVHD) prevention during unrelated-donor hematopoietic cell transplantation (HCT). All patients received cyclosporine/methotrexate plus 4 doses of abatacept (10 mg/kg/dose) on days −1, +5, +14, +28 post-HCT. The feasibility of adding abatacept, its pharmacokinetics, pharmacodynamics, and its impact on aGVHD, infection, relapse, and transplantation-related mortality (TRM) were assessed. All patients received the planned abatacept doses, and no infusion reactions were noted. Compared with a cohort of patients not receiving abatacept (the StdRx cohort), patients enrolled in the study (the ABA cohort) demonstrated significant inhibition of early CD4+ T cell proliferation and activation, affecting predominantly the effector memory (Tem) subpopulation, with 7- and 10-fold fewer proliferating and activated CD4+ Tem cells, respectively, at day+28 in the ABA cohort compared with the StdRx cohort (P
- Published
- 2013
- Full Text
- View/download PDF
43. Evidence for Kidney Rejection After Combined Bone Marrow and Renal Transplantation Despite Ongoing Whole-Blood Chimerism in Rhesus Macaques
- Author
-
F. Leopardi, Kelly Hamby, M. Song, Christian P. Larsen, Taylor Deane, Alton B. Farris, Andrew J. Page, Elizabeth Strobert, Sharon Sen, Linda Stempora, Allan D. Kirk, Leslie S. Kean, Karnail Singh, Aneesah Polnett, and Swetha Ramakrishnan
- Subjects
Graft Rejection ,medicine.medical_treatment ,T cell ,chemical and pharmacologic phenomena ,Chimerism ,Article ,medicine ,Transplantation, Homologous ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,Kidney transplantation ,Bone Marrow Transplantation ,Transplantation ,Kidney ,business.industry ,Immunosuppression ,medicine.disease ,Macaca mulatta ,Kidney Transplantation ,surgical procedures, operative ,medicine.anatomical_structure ,Sirolimus ,Immunology ,Bone marrow ,business ,CD8 ,medicine.drug - Abstract
Although there is evidence linking hematopoietic chimerism induction and solid organ transplant tolerance, the mechanistic requirements for chimerism-induced tolerance are not clearly elucidated. To address this, we used an MHC-defined primate model to determine the impact of impermanent, T cell-poor, mixed-chimerism on renal allograft survival. We compared two cohorts: one receiving a bone marrow and renal transplant ("BMT/renal") and one receiving only a renal transplant. Both cohorts received maintenance immunosuppression with CD28/CD40-directed costimulation blockade and sirolimus. As previously demonstrated, this transplant strategy consistently induced compartmentalized donor chimerism, (significant whole-blood chimerism, lacking T cell chimerism). This chimerism was not sufficient to prolong renal allograft acceptance: the BMT/renal mean survival time (MST, 76 days) was not significantly different than the renal transplant alone MST (85 days, p = 0.46), with histopathology documenting T cell mediated rejection. Flow cytometric analysis revealed significant enrichment for CD28-/CD95+ CD4+ and CD8+ Tem cells in the rejected kidney, suggesting a link between CD28-negative Tem and costimulation blockade-resistant rejection. These results suggest that in some settings, transient T cell-poor chimerism is not sufficient to induce tolerance to a concurrently placed renal allograft and that the presence of this chimerism per se is not an independent biomarker to identify tolerance.
- Published
- 2012
- Full Text
- View/download PDF
44. CD40 Blockade Combines with CTLA4Ig and Sirolimus to Produce Mixed Chimerism in an MHC-Defined Rhesus Macaque Transplant Model
- Author
-
Taylor Deane, Linda Stempora, Andrew J. Page, Keith A. Reimann, Allan D. Kirk, Sharon Sen, Kelly Hamby, Christian P. Larsen, Maria C. Russell, Elizabeth Strobert, Karnail Singh, Leslie S. Kean, F. Leopardi, Swetha Srinivasan, and Andrew A. Price
- Subjects
Immunoconjugates ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Hematopoietic stem cell transplantation ,Chimerism ,Article ,Immune tolerance ,Abatacept ,CD28 Antigens ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,CD40 Antigens ,CD154 ,Sirolimus ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Antibodies, Monoclonal ,CD28 ,hemic and immune systems ,Immunosuppression ,Flow Cytometry ,Macaca mulatta ,Blockade ,Models, Animal ,Immunology ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
In murine models, T-cell costimulation blockade of the CD28:B7 and CD154:CD40 pathways synergistically promotes immune tolerance after transplantation. While CD28 blockade has been successfully translated to the clinic, translation of blockade of the CD154:CD40 pathway has been less successful, in large part due to thromboembolic complications associated with anti-CD154 antibodies. Translation of CD40 blockade has also been slow, in part due to the fact that synergy between CD40 blockade and CD28 blockade had not yet been demonstrated in either primate models or humans. Here we show that a novel, non-depleting CD40 monoclonal antibody, 3A8, can combine with combined CTLA4Ig and sirolimus in a well-established primate bone marrow chimerism-induction model. Prolonged engraftment required the presence of all three agents during maintenance therapy, and resulted in graft acceptance for the duration of immunosuppressive treatment, with rejection resulting upon immunosuppression withdrawal. Flow cytometric analysis revealed that upregulation of CD95 expression on both CD4+ and CD8+ T-cells correlated with rejection, suggesting that CD95 may be a robust biomarker of graft loss. These results are the first to demonstrate prolonged chimerism in primates treated with CD28/mTOR blockade and non-depletional CD40 blockade, and support further investigation of combined costimulation blockade targeting the CD28 and CD40 pathways.
- Published
- 2012
- Full Text
- View/download PDF
45. Pharmacologic Blockade of Notch/Delta-like Ligand 4 Signaling Protects from Gastrointestinal Acute Graft-Versus-Host Disease in Non-Human Primates
- Author
-
Alison Yu, Angela Panoskaltsis-Mortari, Agne Taraseviciute, Steve Carpenter, Olivier Harari, Kayla Betz, Frank Kuhnert, Hengqi Zheng, Victor Tkachev, Lucrezia Colonna, Bruce R. Blazar, Daniel J. Hunt, Judith M Carlson, Virginia Hoglund, Leslie S. Kean, Scott N. Furlan, Gavin Thurston, and Ivan Maillard
- Subjects
0301 basic medicine ,education.field_of_study ,Delta-like ligand 4 ,Regulatory T cell ,business.industry ,T cell ,Immunology ,Notch signaling pathway ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Blockade ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Graft-versus-host disease ,medicine ,Cancer research ,Cytotoxic T cell ,business ,education ,Tissue homeostasis - Abstract
The Notch signaling pathway is an evolutionarily conserved, cell-cell communication system with critical functions in organogenesis and tissue homeostasis, including hemato- and immuno-poiesis. Recent data have revealed important roles for Notch in the regulation of mature T cell differentiation and function. Studies in mouse models have identified Notch as a critical regulator of pathogenic T-cell responses during acute GVHD (aGVHD) (Zhang Y, 2011, Blood). However, the exact biological effects and the therapeutic potential of Notch pathway manipulation in clinical settings remains unclear. To address this question, we tested the activity of Notch pathway blockade in a non-human primate (NHP) aGVHD model, previously shown to exhibit donor T cell-intrinsic activation of the Notch pathway during aGVHD (Furlan SN, 2015, Sci Transl Med). To inhibit the Notch pathway, we used a blocking mAb to the Notch ligand DLL4, identified as the dominant ligand in a mouse aGVHD model (Tran, 2013, JCI; Chung, 2017, JCI). Prophylactic treatment regimens with either a single administration of anti-DLL4 mAb on day 0 (3 mg/kg), or with 3 doses (3 mg/kg each) on days 0, 7 and 14 significantly improved GVHD-free survival of allo-HCT recipients (median survival time (MST) = 26.5 days for the single dose regimen, and MST = 26 days for the triple dose regimen) in comparison with unprophylaxed controls (MST = 8 days, p We next sought to determine the mechanism of GI aGVHD protection caused by DLL4 inhibition. Notably, anti-DLL4 GVHD prophylaxis decreased the expression of the gut-homing α4β7 integrin on CD8 T cells from the peripheral blood, spleen, mesenteric lymph nodes and colon in treated recipients (Fig 2A). Intestinal CD8 T cells also displayed a skewing towards a mature, effector-memory phenotype (Fig 2B) and reduced Ki67+ proliferation (Fig 2C). In contrast, CD8 T cells isolated from liver, the main site of break-through GVHD in anti-DLL4 mAb-treated animals, demonstrated a similar proliferation rate in anti-DLL4-treated compared to unprophylaxed cohorts (Fig 2C). Moreover, anti-DLL4 mAb prophylaxis was associated with an improved regulatory T cell/conventional T cell ratio in the colon but not in liver (Fig 2D). Collectively, our results show that the function of individual Notch ligands is preserved from mice to primates in aGVHD. We provide the first demonstration of the important role of Notch signaling in alloimmune T cell activation in a large animal model of aGVHD, and the therapeutic potential of DLL4 blockade for aGVHD prevention. Disclosures Tkachev: Regeneron Pharmaceuticals, Inc.: Research Funding. Kuhnert:Regeneron Pharmaceuticals, Inc.: Employment. Blazar:Kadmon Corporation, LLC: Consultancy, Research Funding. Carpenter:Regeneron Pharmaceuticals, Inc.: Employment. Harari:Regeneron Pharmaceuticals, Inc.: Employment. Thurston:Regeneron Pharmaceuticals, Inc.: Employment, Equity Ownership. Kean:Regeneron Pharmaceuticals, Inc.: Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
46. An MHC-Defined Primate Model Reveals Significant Rejection of Bone Marrow After Mixed Chimerism Induction Despite Full MHC Matching
- Author
-
Sharon Sen, Christian P. Larsen, Elizabeth Strobert, Kelly Hamby Linzie, David H. O’Connor, Andrew J. Page, Thea Ward, Karnail Singh, Maria C. Russell, Maria Cecilia T. Penedo, Leslie S. Kean, Weston P. Miller, Linda Stempora, Roger W. Wiseman, and Taylor Deane
- Subjects
Graft Rejection ,Male ,Transplantation Conditioning ,T-Lymphocytes ,T cell ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Article ,Immune tolerance ,Major Histocompatibility Complex ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,Busulfan ,Immunosuppression Therapy ,Sirolimus ,Transplantation Chimera ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Immunosuppression ,medicine.disease ,Macaca mulatta ,Substance Withdrawal Syndrome ,Transplant rejection ,Tolerance induction ,surgical procedures, operative ,medicine.anatomical_structure ,Histocompatibility ,Immunology ,Female ,Bone marrow ,business ,medicine.drug - Abstract
In murine models, mixed hematopoietic chimerism induction leads to robust immune tolerance. However, translation to primates and to patients has been difficult. In this study, we used a novel MHC-defined rhesus macaque model to examine the impact of MHC matching on the stability of costimulation blockade-/sirolimus-mediated chimerism, and to probe possible mechanisms of bone marrow rejection after nonmyeloablative transplant. Using busulfan-based pretransplant preparation and maintenance immunosuppression with sirolimus, as well as CD28 and CD154 blockade, all recipients demonstrated donor engraftment after transplant. However, the mixed chimerism that resulted was compartmentalized, with recipients demonstrating significantly higher whole blood chimerism compared to T cell chimerism. Thus, the vast majority of T cells presenting posttransplant were recipient-rather than donor-derived. Surprisingly, even in MHC-matched transplants, rejection of donor hematopoiesis predominated after immunosuppression withdrawal. Weaning of immunosuppression was associated with a surge of antigen-experienced T cells, and transplant rejection was associated with the acquisition of donor-directed T cell alloreactivity. These results suggest that a reservoir of alloreactive cells was present despite prior costimulation blockade and sirolimus, and that the post-immunosuppression lymphocytic rebound may have lead to a phenotypic shift in these recipient T cells towards an activated, antigen-experienced phenotype, and ultimately, to transplant rejection.
- Published
- 2010
- Full Text
- View/download PDF
47. Induction of Chimerism in Rhesus Macaques through Stem Cell Transplant and Costimulation Blockade-Based Immunosuppression
- Author
-
Christian P. Larsen, Leslie S. Kean, J. Jiang, Megan M. Durham, Elizabeth Strobert, Kenneth Cardona, Kelly Hamby, Thomas R. Jones, Mark R. Rigby, Thomas C. Pearson, Daniel G. Anderson, Shivaprakash Gangappa, Nozomu Shirasugi, H. Bello, Rose Hendrix, and Andrew B. Adams
- Subjects
T-Lymphocytes ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Chimerism ,medicine ,Animals ,Immunology and Allergy ,Pharmacology (medical) ,Leukapheresis ,Busulfan ,Bone Marrow Transplantation ,Immunosuppression Therapy ,Sirolimus ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Receptors, Interleukin-2 ,Immunosuppression ,Macaca mulatta ,Blockade ,Haematopoiesis ,medicine.anatomical_structure ,Cytomegalovirus Infections ,Immunology ,Transplantation Tolerance ,Bone marrow ,Stem cell ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
A strategy for producing high-level hematopoietic chimerism after non-myeloablative conditioning has been established in the rhesus macaque. This strategy relies on hematopoietic stem cell transplantation after induction with a non-myeloablative dose of busulfan and blockade of the IL2-receptor in the setting of mTOR inhibition with sirolimus and combined CD28/CD154 costimulation blockade. Hematopoietic stem cells derived from bone marrow and leukopheresis products both were found to be successful in inducing high-level chimerism. Mean peripheral blood peak donor chimerism was 81% with a median chimerism duration of 145 days. Additional immune modulation strategies, such as pre-transplant CD8 depletion, donor-specific transfusion, recipient thymectomy or peritransplant deoxyspergualin treatment did not improve the level or durability of chimerism. Recipient immunologic assessment suggested that chimerism occurred amidst donor-specific down-regulation of alloreactive T cells, and the reappearance of vigorous T-mediated alloreactivity accompanied rejection of the transplants. Furthermore, viral reactivation constituted a significant transplant-related toxicity and may have negatively impacted the ability to achieve indefinite survival of transplanted stem cells. Nevertheless, this chimerism-induction regimen induced amongst the longest-lived stem cell chimerism reported to date for non-human primates and thus represents a platform upon which to evaluate emerging tolerance-induction strategies.
- Published
- 2007
- Full Text
- View/download PDF
48. Red Blood Cell Transfusions Are Associated with HLA Class I but not H-Y Alloantibodies in Children with Sickle Cell Disease
- Author
-
Jeanne E. Hendrickson, Howard M. Gebel, Robert A. Bray, Leslie S. Kean, David B. Miklos, Marianne M. Yee, Robert Sheppard Nickel, and John T. Horan
- Subjects
Male ,Adolescent ,H-Y Antigen ,Human leukocyte antigen ,Anemia, Sickle Cell ,Article ,Immunophenotyping ,Young Adult ,Antigen ,Isoantibodies ,Minor histocompatibility antigen ,Odds Ratio ,Medicine ,Humans ,Platelet ,Child ,biology ,business.industry ,Histocompatibility Antigens Class I ,Hematology ,Odds ratio ,Transplantation ,Red blood cell ,medicine.anatomical_structure ,Cross-Sectional Studies ,Child, Preschool ,Immunology ,biology.protein ,Female ,Antibody ,business ,Erythrocyte Transfusion - Abstract
Blood transfusions can induce alloantibodies to antigens on red blood cells (RBCs), white blood cells and platelets, with these alloantibodies affecting transfusion and transplantation. While transfusion-related alloimmunization against RBC antigens and human leucocyte antigens (HLA) have been studied, transfusion-related alloimmunization to minor histocompatibility antigens (mHA), such as H-Y antigens, has not been clinically characterized. We conducted a cross-sectional study of 114 children with sickle cell disease (SCD) and tested for antibodies to 5 H-Y antigens and to HLA class I and class II. Few patients had H-Y antibodies, with no significant differences in the prevalence of any H-Y antibody observed among transfused females (7%), transfused males (6%) and never transfused females (4%). In contrast, HLA class I, but not HLA class II, antibodies were more prevalent among transfused than never transfused patients (class I: 33% vs. 13%, P = 0·046; class II: 7% vs. 8%, P = 0·67). Among transfused patients, RBC alloantibody history but not amount of transfusion exposure was associated with a high (25%) HLA class I panel reactive antibody (Odds ratio 6·8, 95% confidence interval 2·1-22·3). These results are consistent with immunological responder and non-responder phenotypes, wherein a subset of patients with SCD may be at higher risk for transfusion-related alloimmunization.
- Published
- 2015
49. NK Cells Mediate Costimulation Blockade-Resistant Rejection of Allogeneic Stem Cells During Nonmyeloablative Transplantation
- Author
-
Eun D.Han Lee, Thomas C. Pearson, Brent H. Koehn, Christian P. Larsen, Kelly Hamby, Andrew B. Adams, Linda Stempora, Leslie S. Kean, E A Heiss, and Shana M. Coley
- Subjects
Graft Rejection ,medicine.medical_treatment ,Natural killer cell ,Immune tolerance ,Mice ,Interleukin 21 ,medicine ,Animals ,Transplantation, Homologous ,Immunology and Allergy ,Pharmacology (medical) ,Bone Marrow Transplantation ,Mice, Inbred BALB C ,Transplantation ,CD40 ,biology ,business.industry ,Killer Cells, Natural ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,biology.protein ,Stem cell ,business ,Stem Cell Transplantation ,Allotransplantation - Abstract
Although T-cell CD28/CD40 costimulation blockade represents a powerful mechanism to promote immune tolerance during murine allotransplantation, it has not yet been successfully translated to clinical transplantation. We determined the impact of natural killer (NK) cells on costimulation blockade-resistant rejection of donor bone marrow. We found that NK cells represent a potent barrier to engraftment: host NK depletion led to increased donor stem cell survival, increased mixed hematopoietic chimerism and to engraftment of low doses of donor marrow (1 x 10(8)/kg) that were otherwise rejected. To understand the mechanisms of NK alloreactivity, we employed an in vivo NK-specific cytotoxicity assay. We found that an increased proportion of target cells were killed between days 2 and 8 after cell transfer, and that NK killing of parental targets was inducible: NK cells preprimed with allotargets were more efficient at their elimination upon reexposure. Finally, both transplant and in vivo NK-killing models were used to determine the contribution of LFA-1 to NK alloreactivity. Blockade of LFA-1 led to decreased NK-mediated killing, and increased alloengraftment. These results identify NK alloreactivity as an integral component to costimulation blockade-resistant rejection, and suggest that its inhibition may represent an important target in the clinical translation of tolerance-induction transplantation.
- Published
- 2006
- Full Text
- View/download PDF
50. Murine and Math Models for the Level of Stable Mixed Chimerism to Cure β-Thalassemia by Nonmyeloablative Bone Marrow Transplantation
- Author
-
Leslie S. Kean, Lewis L. Hsu, Carla Roberts, David R. Archer, and Can Balkan
- Subjects
Hemolytic anemia ,Transplantation Conditioning ,Thalassemia ,Congenic ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Leukocyte Count ,Mice ,History and Philosophy of Science ,Animals, Congenic ,Animals ,Humans ,Medicine ,Busulfan ,Survival analysis ,Bone Marrow Transplantation ,Transplantation Chimera ,business.industry ,General Neuroscience ,Graft Survival ,Remission Induction ,beta-Thalassemia ,medicine.disease ,Survival Analysis ,Hematopoiesis ,Mice, Inbred C57BL ,Transplantation ,Haematopoiesis ,Immunology ,Erythrocyte Count ,Stem cell ,business ,medicine.drug - Abstract
Stable mixed chimeric stem cell transplantation in hemoglobinopathies exploits shorter erythroid survival in hemolytic anemias, providing normal donor red blood cells with a competitive survival advantage. This study examined the level of stable mixed chimerism necessary for complete hematological cure of the thalassemic phenotype, using a nonmyeloablative busulfan chemotherapeutic preparation. Thalassemic mice transplanted from congenic wild-type donors developed partial mixed chimerism. Hematologic cure required >80% donor red blood cells and only >13% donor white blood cells. Murine and human transplant results were compared with a math model for survival advantage of donor peripheral blood cells produced by steady-state chimeric marrow.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.