197 results on '"Robert Lowsky"'
Search Results
2. Bendamustine is a safe and effective lymphodepletion agent for axicabtagene ciloleucel in patients with refractory or relapsed large B-cell lymphoma
- Author
-
Crystal Mackall, Holden T Maecker, Lekha Mikkilineni, Melody Smith, Saurabh Dahiya, Bita Sahaf, David Miklos, Sally Arai, Surbhi Sidana, Wen-Kai Weng, Dasom Lee, Anmol Goyal, Sushma Bharadwaj, Eric Lau, Mark P Hamilton, Hrishi Srinagesh, Alexandria Jensen, Jayasindhu Mallampet, Sarah Elkordy, Shriya Syal, Sunita Patil, Theresa Latchford, Laura J Johnston, Robert Lowsky, Robert Negrin, Andrew R Rezvani, Judith Shizuru, Everett H Meyer, Parveen Shiraz, Lori Muffly, and Matthew J Frank
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Fludarabine in combination with cyclophosphamide (FC) is the standard lymphodepletion regimen for CAR T-cell therapy (CAR T). A national fludarabine shortage in 2022 necessitated the exploration of alternative regimens with many centers employing single-agent bendamustine as lymphodepletion despite a lack of clinical safety and efficacy data. To fill this gap in the literature, we evaluated the safety, efficacy, and expansion kinetics of bendamustine as lymphodepletion prior to axicabtagene ciloleucel (axi-cel) therapy.Methods 84 consecutive patients with relapsed or refractory large B-cell lymphoma treated with axi-cel and managed with a uniform toxicity management plan at Stanford University were studied. 27 patients received alternative lymphodepletion with bendamustine while 57 received FC.Results Best complete response rates were similar (73.7% for FC and 74% for bendamustine, p=0.28) and there was no significant difference in 12-month progression-free survival or overall survival estimates (p=0.17 and p=0.62, respectively). The frequency of high-grade cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome was similar in both the cohorts. Bendamustine cohort experienced lower proportions of hematological toxicities and antibiotic use for neutropenic fever. Immune reconstitution, as measured by quantitative assessment of cellular immunity, was better in bendamustine cohort as compared with FC cohort. CAR T expansion as measured by peak expansion and area under the curve for expansion was comparable between cohorts.Conclusions Bendamustine is a safe and effective alternative lymphodepletion conditioning for axi-cel with lower early hematological toxicity and favorable immune reconstitution.
- Published
- 2024
- Full Text
- View/download PDF
3. Establishment of Chimerism and Organ Transplant Tolerance in Laboratory Animals: Safety and Efficacy of Adaptation to Humans
- Author
-
Robert Lowsky and Samuel Strober
- Subjects
chimerism ,transplant tolerance ,immune suppression ,animal models ,human trials ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The definition of immune tolerance to allogeneic tissue and organ transplants in laboratory animals and humans continues to be the acceptance of the donor graft, rejection of third-party grafts, and specific unresponsiveness of recipient immune cells to the donor alloantigens in the absence of immunosuppressive treatments. Actively acquired tolerance was achieved in mice more than 60 years ago by the establishment of mixed chimerism in neonatal mice. Once established, mixed chimerism was self-perpetuating and allowed for acceptance of tissue transplants in adults. Successful establishment of tolerance in humans has now been reported in several clinical trials based on the development of chimerism after combined transplantation of hematopoietic cells and an organ from the same donor. This review examines the mechanisms of organ graft acceptance after establishment of mixed chimerism (allo-tolerance) or complete chimerism (self-tolerance), and compares the development of graft versus host disease (GVHD) and graft versus tumor (GVT) activity in complete and mixed chimerism. GVHD, GVT activity, and complete chimerism are also discussed in the context of bone marrow transplantation to treat hematologic malignancies. The roles of transient versus persistent mixed chimerism in the induction and maintenance of tolerance and organ graft acceptance in animal models and clinical studies are compared. Key differences in the stability of mixed chimeras and tolerance induction in MHC matched and mismatched rodents, large laboratory animals, and humans are examined to provide insights into the safety and efficacy of translation of results of animal models to clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
4. Nonmyeloablative TLI-ATG conditioning for allogeneic transplantation: mature follow-up from a large single-center cohort
- Author
-
Michael A. Spinner, Vanessa E. Kennedy, John S. Tamaresis, Philip W. Lavori, Sally Arai, Laura J. Johnston, Everett H. Meyer, David B. Miklos, Lori S. Muffly, Robert S. Negrin, Andrew R. Rezvani, Judith A. Shizuru, Wen-Kai Weng, Richard T. Hoppe, Samuel Strober, and Robert Lowsky
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Nonmyeloablative total lymphoid irradiation and antithymocyte globulin (TLI-ATG) conditioning is protective against graft-versus-host disease (GVHD), while retaining graft-versus-tumor activity across various hematologic malignancies. We report our comprehensive experience using TLI-ATG conditioning in 612 patients with hematologic malignancies who underwent allogeneic transplantation at Stanford University from 2001 to 2016. All patients received granulocyte colony-stimulating factor–mobilized peripheral blood grafts and cyclosporine and mycophenolate mofetil for GVHD prophylaxis. The median age was 60 years (range, 21-78), with a median follow-up of 6.0 years (range, 1.0-16.4). Common diagnoses included acute myeloid leukemia (AML; n = 193), myelodysplastic syndrome (MDS; n = 94), chronic lymphocytic leukemia (CLL; n = 80), non-Hodgkin lymphoma (NHL; n = 175), and Hodgkin lymphoma (HL; n = 35). Thirty-four percent of patients had a comorbidity index ≥3, 30% had a high to very high disease risk index, and 56% received unrelated donor grafts, including 15% with HLA-mismatched donors. Ninety-eight percent underwent transplant in the outpatient setting, and 57% were never hospitalized from days 0 through 100. The 1-year rates of nonrelapse mortality (NRM), grade II-IV acute GVHD, and extensive chronic GVHD were 9%, 14%, and 22%, respectively. The 4-year estimates for overall and progression-free survival were 42% and 32% for AML, 30% and 21% for MDS, 67% and 43% for CLL, 68% and 45% for NHL, and 78% and 49% for HL. Mixed chimerism correlated with the risk of relapse. TLI-ATG conditioning was well tolerated, with low rates of GVHD and NRM. Durable remissions were observed across hematologic malignancies, with particularly favorable outcomes for heavily pretreated lymphomas. Several efforts are underway to augment donor chimerism and reduce relapse rates while maintaining the favorable safety and tolerability profile of this regimen.
- Published
- 2019
- Full Text
- View/download PDF
5. Allogeneic transplantation using TLI-ATG conditioning for Hodgkin lymphoma after failure of autologous transplantation
- Author
-
Michael A. Spinner, Ranjana H. Advani, Richard T. Hoppe, Robert Lowsky, and Lori S. Muffly
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2018
- Full Text
- View/download PDF
6. Infusion of donor-derived CD8+ memory T cells for relapse following allogeneic hematopoietic cell transplantation
- Author
-
Lori Muffly, Kevin Sheehan, Randall Armstrong, Kent Jensen, Keri Tate, Andrew R. Rezvani, David Miklos, Sally Arai, Judith Shizuru, Laura Johnston, Everett Meyer, Wen-Kai Weng, Ginna G. Laport, Robert S. Negrin, Sam Strober, and Robert Lowsky
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Murine models showed that CD8+CD44hi memory T (TM) cells could eradicate malignant cells without inducing graft-versus-host disease (GVHD). We evaluated the feasibility and safety of infusing freshly isolated and purified donor-derived phenotypic CD8+ TM cells into adults with disease relapse after allogeneic hematopoietic cell transplantation (HCT). Phenotypic CD8 TM cells were isolated after unmobilized donor apheresis using a tandem immunomagnetic selection strategy of CD45RA depletion followed by CD8+ enrichment. Fifteen patients received CD8+ TM cells at escalating doses (1 × 106, 5 × 106, or 10 × 106 cells per kg). Thirteen received cytoreduction before CD8+ TM cell infusion, and 9 had active disease at the time of infusion. Mean yield and purity of the CD8+ TM infusion were 38.1% and 92.8%, respectively; >90% had CD8+ T effector memory phenotype, cytokine expression, and secretion profile. No adverse infusional events or dose-limiting toxicities occurred; GVHD developed in 1 patient (grade 2 liver). Ten patients (67%) maintained or achieved response (7 complete response, 1 partial response, 2 stable disease) for at least 3 months after infusion; 4 of the responders had active disease at the time of infusion. With a median follow-up from infusion of 328 days (range, 118-1328 days), median event-free survival and overall survival were 4.9 months (95% confidence interval [CI], 1-19.3 months) and 19.6 months (95% CI, 5.6 months to not reached), respectively. Collection and enrichment of phenotypic CD8+ TM cells is feasible, well tolerated, and associated with a low incidence of GVHD when administered as a manipulated infusion of donor lymphocytes in patients who have relapsed after HCT. This trial was registered at www.clinicaltrials.gov as #NCT01523223.
- Published
- 2018
- Full Text
- View/download PDF
7. HLA-mismatched unrelated donor transplantation using TLI-ATG conditioning has a low risk of GVHD and potent antitumor activity
- Author
-
Michael A. Spinner, Marcelo Fernández-Viña, Lisa E. Creary, Olivia Quinn, Linda Elder, Sally Arai, Laura J. Johnston, Everett H. Meyer, David B. Miklos, Lori S. Muffly, Robert S. Negrin, Judith A. Shizuru, Wen-Kai Weng, Ginna G. Laport, Samuel Strober, Robert Lowsky, and Andrew R. Rezvani
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Many patients lack a fully HLA-matched donor for hematopoietic cell transplantation (HCT), and HLA mismatch is typically associated with inferior outcomes. Total lymphoid irradiation and antithymocyte globulin (TLI-ATG) is a nonmyeloablative conditioning regimen that is protective against graft-versus-host disease (GVHD), and we hypothesized that the protective effect would extend beyond HLA-matched donors. We report outcomes for all consecutively transplanted patients at Stanford University from December 2001 through May 2015 who received TLI-ATG conditioning and HCTs from 8 to 9 out of 10 HLA-mismatched unrelated donors (MMUDs, N = 72) compared with 10 out of 10 HLA-matched unrelated donors (MUDs, N = 193). The median age of the patients was 60 years with a median follow-up of 2 years, and there was a similar distribution of lymphoid and myeloid malignancies in both cohorts. There were no significant differences between MMUD and MUD cohorts in overall survival (46% vs 46% at 5 years, P = .86), disease-free survival (38% vs 28% at 5 years, P = .25), nonrelapse mortality (17% vs 12% at 2 years, P = .34), acute GVHD grades III-IV (6% vs 3% at day +100, P = .61), or chronic GVHD (39% vs 35% at 5 years, P = .49). There was a trend toward less relapse in the MMUD cohort (45% vs 60% at 5 years, hazard ratio: 0.71, P = .094), which was significant for patients with lymphoid malignancies (29% vs 57% at 5 years, hazard ratio: 0.55, P = .044). Achieving full donor chimerism was strongly associated with lower relapse rates. TLI-ATG conditioning may overcome the traditionally poorer outcome associated with HLA-mismatched donors and may be particularly well suited for patients with lymphoid malignancies who lack HLA-matched donors.
- Published
- 2017
- Full Text
- View/download PDF
8. Erratum: Pregnancy Complicated by Gorham–Stout Disease and Refractory Chylothorax
- Author
-
Jessica Hellyer, Hunter Oliver-Allen, Majid Shafiq, Alisha Tolani, Maurice Druzin, Michael Jeng, Stanley Rockson, and Robert Lowsky
- Subjects
Gynecology and obstetrics ,RG1-991 - Published
- 2016
- Full Text
- View/download PDF
9. Pregnancy Complicated by Gorham–Stout Disease and Refractory Chylothorax
- Author
-
Jessica Hellyer, Hunter Oliver-Allen, Majid Shafiq, Alisha Tolani, Maurice Druzin, Michael Jeng, Stanley Rockson, and Robert Lowsky
- Subjects
pregnancy ,chylothorax ,gorham–stout disease ,total parenteral nutrition ,octreotide ,Gynecology and obstetrics ,RG1-991 - Abstract
Abstract Introduction Gorham–Stout Disease (GSD) is a rare disorder of bony destruction due to lymphangiomatosis, and is often triggered by hormones. One complication of GSD is the development of chylothorax, which carries a high mortality rate. Very little experience has been published to guide management in GSD during pregnancy to optimize both fetal and maternal health. Case Study A 20-year-old woman with known GSD presented with shortness of breath at 18 weeks of pregnancy, due to bilateral chylothoraces which required daily drainage. To minimize chylous fluid formation, she was placed on bowel rest with total parenteral nutrition (limiting lipid intake) and received octreotide to decrease splanchnic blood flow and chylous fluid drainage. Treatment options were limited due to her pregnancy. Twice daily home chest tube drainage of a single lung cavity, total parenteral nutrition, octreotide, and albumin infusions allowed successful delivery of a healthy 37 weeks' gestation infant by cesarean delivery. Discussion This case illustrates the management of a rare clinical disease of bone resorption and lymphangiomatosis complicated by bilateral, refractory chylothoraces, triggered by pregnancy, in whom treatment options are limited, and the need for a multidisciplinary health care team to ensure successful maternal and fetal outcomes.
- Published
- 2016
- Full Text
- View/download PDF
10. Risks and benefits of sex-mismatched hematopoietic cell transplantation differ according to conditioning strategy
- Author
-
Hideki Nakasone, Mats Remberger, Lu Tian, Petter Brodin, Bita Sahaf, Fang Wu, Jonas Mattsson, Robert Lowsky, Robert Negrin, David B. Miklos, and Everett Meyer
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Sex-mismatched hematopoietic cell transplantation is linked to increased graft-versus-host disease and mortality in myeloablative conditioning. Here we evaluated outcomes of 1,041 adult transplant recipients at two centers between 2006 and 2013 and investigated how the effect of sex-mismatching differed in myeloablative, reduced-intensity, and non-myeloablative total lymphoid irradiation with anti-thymocyte globulin conditioning. Among patients who underwent myeloablative conditioning, male recipients with female donors had increased chronic graft-versus-host disease (hazard ratio 1.83, P
- Published
- 2015
- Full Text
- View/download PDF
11. Improved outcomes for relapsed/refractory Hodgkin lymphoma after autologous transplantation in the era of novel agents
- Author
-
Michael A Spinner, R. Alejandro Sica, John S Tamaresis, Ying Lu, Cheryl Chang, Robert Lowsky, Matthew J. Frank, Laura J Johnston, David B Miklos, Lori Muffly, Robert S. Negrin, Andrew R. Rezvani, Parveen Shiraz, Judith A. Shizuru, Wen-Kai Weng, Michael S. Binkley, Richard T. Hoppe, Ranjana H Advani, and Sally Arai
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
The treatment landscape of relapsed/refractory (R/R) classic Hodgkin lymphoma (cHL) has evolved significantly over the past decade following the approval of brentuximab vedotin (BV) and the programmed death-1 (PD-1) inhibitors. We evaluated how outcomes and practice patterns have changed for R/R cHL patients who underwent autologous hematopoietic cell transplantation (AHCT) at our institution from 2011-2020 (N=183) compared to 2001-2010 (N=159) and evaluated prognostic factors for progression-free survival (PFS) and overall survival (OS) in both eras. OS was superior in the modern era (4-year estimates 89.1% vs 79.0%, HR 0.53, 95% CI 0.33-0.85, p=0.011) with a trend towards lower non-relapse mortality beyond 2 years post-transplant. Among patients who progressed after AHCT, 4-year post-progression survival increased from 43.3% to 71.4% in the modern era, reflecting increasing use of BV and the PD-1 inhibitors. In multivariable analysis for patients transplanted in the modern era, age ³45 years, primary refractory disease, and lack of complete remission pre-AHCT were associated with inferior PFS, while receipt of a PD-1 inhibitor-based regimen pre-AHCT was associated with superior PFS (HR 0.21, 95% CI 0.05-0.80, p=0.030). Extranodal disease at relapse was associated with inferior OS (HR 3.12, 95% CI 1.25-7.77, p=0.014). Our study demonstrates improved survival for R/R cHL after AHCT in the modern era attributed to more effective salvage regimens allowing for better disease control pre-AHCT and improved outcomes for patients who progressed after AHCT. Excellent outcomes were observed with PD-1 inhibitor-based salvage regimens pre-AHCT and support a randomized trial evaluating immunotherapy in the second line setting.
- Published
- 2023
12. Outcomes for Acute Myeloid Leukemia Relapse after Allogeneic Hematopoietic Cell Transplantation Remain Poor in the Modern Era
- Author
-
Shona Philip, Robert Lowsky, Laura J. Johnston, Sally Arai, Everett H. Meyer, Robert S. Negrin, Andrew R. Rezvani, Parveen Shiraz, Judith A. Shizuru, Surbhi Sidana, Wen-Kai Weng, Sushma Bharadwaj, Matthew J. Frank, David B. Miklos, Melody Smith, Lori Muffly, and Vaibhav Agrawal
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
13. The Development of Carhlh after Axicabtagene Ciloleucel Is Associated with Poor Outcomes
- Author
-
Shona Philip, Hrishikesh K. Srinagesh, Mark P Hamilton, Cesar Gentille, Alain Mina, Sally Arai, Laura J. Johnston, Robert Lowsky, Everett H Meyer, Robert S. Negrin, Andrew R. Rezvani, Parveen Shiraz, Judith A Shizuru, Surbhi Sidana, Wen-Kai Weng, Sushma Bharadwaj, Saurabh Dahiya, Lori Muffly, Melody Smith, David B. Miklos, and Matthew J Frank
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
14. Analysis of Bendamustine Lymphodepletion, CD19 CART Expansion, Safety and Efficacy in Patients with Rel/Ref Non-Hodgkin Lymphoma
- Author
-
Sushma Bharadwaj, Mark P Hamilton, Bita Sahaf, John Tamaresis, Sunita Patil, Paul J Hanson, Theresa Latchford, Sally Arai, Laura J. Johnston, Robert Lowsky, Robert S. Negrin, Andrew R. Rezvani, Judith A Shizuru, Everett H Meyer, Parveen Shiraz, Surbhi Sidana, Melody Smith, Wen-Kai Weng, Lori Muffly, Crystal L. Mackall, Matthew J Frank, David B. Miklos, and Saurabh Dahiya
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
15. Precision-Engineered Cell Therapy Orca-T Demonstrates High Relapse-Free Survival at 1 Year While Reducing Graft-Versus-Host Disease and Toxicity
- Author
-
Caspian Oliai, Rasmus T Hoeg, Anna Pavlova, Arpita Gandhi, Lori Muffly, Rohtesh S. Mehta, Samer A Srour, Edmund K. Waller, Robert Lowsky, Sagar S. Patel, Bhagirathbhai Dholaria, Carlos Bachier, Jeremy M. Pantin, Amandeep Salhotra, Joseph P. McGuirk, Nathaniel B Fernhoff, J Scott McClellan, Mehrdad Abedi, Robert S. Negrin, and Everett H Meyer
- Subjects
Transplantation ,Immunology ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
16. Outcomes of adults with lymphoma treated with nonmyeloablative TLI-ATG and radiation boost to high risk or residual disease before allogeneic hematopoietic cell transplant
- Author
-
Michael Dworkin, R. Advani, Susan M. Hiniker, A L Jiang, Michael A. Spinner, Robert Lowsky, R. Von Eyben, and Richard T. Hoppe
- Subjects
Transplantation ,medicine.medical_specialty ,Mycosis fungoides ,Hematopoietic cell ,business.industry ,Hematology ,Disease ,medicine.disease ,Gastroenterology ,Lymphoma ,Lesion ,Extranodal Disease ,Time to recurrence ,Internal medicine ,Toxicity ,medicine ,medicine.symptom ,business - Abstract
We evaluated the impact on survival of antithymocyte globulin conditioning (TLI-ATG) with radiation (RT) boost to high risk or residual disease before allogeneic hematopoietic cell transplant (allo-HCT) for adults with lymphoma (excluding mycosis fungoides and low-grade NHL other than SLL/CLL). Of 251 evaluable patients, 36 received an RT boost within 3 months of allo-HCT at our institution from 2001 to 2016. At the time of TLI-ATG, patients who received boost vs no boost had a lower rate of CR (11% vs 47%, p = 0.0003), higher rates of bulky disease (22% vs 4%, p < 0.0001), extranodal disease (39% vs 5%, p < 0.0001), and positive PET (75% vs 28%, p < 0.00001). In the boost group, the median (range) largest axial lesion diameter was 5.2 cm (1.8-22.3). Median follow-up was 50.2 months (range: 1-196). There was no significant difference in OS, time to recurrence, or time to graft failure with vs without boost. A trend toward higher percent donor CD3+ chimerism was seen with vs without boost (p = 0.0819). The worst boost-related toxicity was grade 2 dermatitis. RT boost may help successfully mitigate the risk of high risk or clinically evident residual disease in adults with lymphoma undergoing allo-HCT.
- Published
- 2021
17. Concordance of peripheral blood and bone marrow measurable residual disease in adult acute lymphoblastic leukemia
- Author
-
Michaela Liedtke, Eric Kuo, David B. Miklos, Ilana R. Yurkiewicz, Matthew J. Frank, Sally Arai, Andrew R. Rezvani, Judith A. Shizuru, Everett Meyer, Laura Johnston, Surbhi Sidana, Sarah Burnash, Hyma T. Vempaty, Wen-Kai Weng, Vandana Sundaram, Connie Chen, Parveen Shiraz, Lori Muffly, Robert S. Negrin, Robert Lowsky, and Jay Y. Spiegel
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,medicine.medical_treatment ,Concordance ,Hematopoietic stem cell transplantation ,Bone Marrow ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Neoplasm ,Prospective Studies ,Prospective cohort study ,medicine.diagnostic_test ,business.industry ,Hematopoietic Stem Cell Transplantation ,Bone Marrow Examination ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Stimulus Report ,body regions ,Bone marrow examination ,Transplantation ,medicine.anatomical_structure ,Adult Acute Lymphoblastic Leukemia ,Bone marrow ,business - Abstract
Monitoring of measurable residual disease (MRD) is essential to the management of acute lymphoblastic leukemia (ALL) and is typically performed through repeated bone marrow (BM) assessments. Using a next-generation sequencing (NGS) MRD platform, we performed a prospective observational study evaluating the correlation between peripheral blood (PB) and BM MRD in adults with ALL receiving cellular therapies (hematopoietic cell transplantation [HCT] and chimeric antigen receptor T-cell [CAR-T] therapies). Among the study cohort (N = 69 patients; 126 paired PB/BM samples), we found strong correlation between PB and BM MRD (r = 0.87; P < .001), with a sensitivity and specificity of MRD detection in the PB of 87% and 90%, respectively, relative to MRD in the BM. MRD became detectable in the PB in 100% of patients who subsequently relapsed following HCT, with median time from MRD+ to clinical relapse of 90 days, and in 85% of patients who relapsed following CAR T, with median time from MRD+ to clinical relapse of 60 days. In adult patients with ALL undergoing cellular therapies, we demonstrate strong concordance between NGS-based MRD detected in the PB and BM. Monitoring of ALL MRD in the PB appears to be an adequate alternative to frequent invasive BM evaluations in this clinical setting.
- Published
- 2021
18. CD22-directed CAR T-cell therapy induces complete remissions in CD19-directed CAR–refractory large B-cell lymphoma
- Author
-
Warren D. Reynolds, Bita Sahaf, Zachary Ehlinger, Steven A. Feldman, Matthew J. Frank, Andrew R. Rezvani, Surbhi Sidana, Jay Y. Spiegel, Allison P. Jacob, Lori Muffly, Kara L. Davis, Shabnum Patel, Jean Oak, Eric H. Yang, Ilan R. Kirsch, Robert Lowsky, Laura Johnston, Juliana Craig, John S. Tamaresis, Michael G. Ozawa, John H. Baird, Sheren F. Younes, David B. Miklos, Parveen Shiraz, Chelsea D. Mullins, Crystal L. Mackall, Robert S. Negrin, Liora M. Schultz, Wen-Kai Weng, Everett Meyer, Sally Arai, Yasodha Natkunam, and Sneha Ramakrishna
- Subjects
Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Sialic Acid Binding Ig-like Lectin 2 ,Antigens, CD19 ,Immunology ,Immunotherapy, Adoptive ,Biochemistry ,CD19 ,Refractory ,Internal medicine ,medicine ,Humans ,Adverse effect ,B-cell lymphoma ,Clinical Trials, Phase I as Topic ,biology ,business.industry ,Remission Induction ,CD22 ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Chimeric antigen receptor ,Lymphoma ,biology.protein ,Chimeric Antigen Receptor T-Cell Therapy ,Lymphoma, Large B-Cell, Diffuse ,business - Abstract
The prognosis of patients with large B-cell lymphoma (LBCL) that progresses after treatment with chimeric antigen receptor (CAR) T-cell therapy targeting CD19 (CAR19) is poor. We report on the first 3 consecutive patients with autologous CAR19-refractory LBCL who were treated with a single infusion of autologous 1 × 106 CAR+ T cells per kilogram targeting CD22 (CAR22) as part of a phase 1 dose-escalation study. CAR22 therapy was relatively well tolerated, without any observed nonhematologic adverse events higher than grade 2. After infusion, all 3 patients achieved complete remission, with all responses continuing at the time of last follow-up (mean, 7.8 months; range, 6-9.3). Circulating CAR22 cells demonstrated robust expansion (peak range, 85.4-350 cells per microliter), and persisted beyond 3 months in all patients with continued radiographic responses and corresponding decreases in circulating tumor DNA beyond 6 months after infusion. Further accrual at a higher dose level in this phase 1 dose-escalation study is ongoing and will explore the role of this therapy in patients in whom prior CAR T-cell therapies have failed. This trial is registered on clinicaltrials.gov as #NCT04088890.
- Published
- 2021
19. Immune reconstitution and infectious complications following axicabtagene ciloleucel therapy for large B-cell lymphoma
- Author
-
Zachary Ehlinger, Everett Meyer, David B. Miklos, Matthew J. Frank, John S. Tamaresis, Laura Johnston, Janice W Brown, Surbhi Sidana, Robert S. Negrin, David J Epstein, John H. Baird, Theresa Latchford, Andrew R. Rezvani, Robert Lowsky, Juliana Craig, Lori Muffly, Sally Arai, Gursharan K. Claire, Wen-Kai Weng, Bita Sahaf, Parveen Shiraz, Crystal L. Mackall, and Jay Y. Spiegel
- Subjects
medicine.medical_specialty ,Clinical Trials and Observations ,medicine.medical_treatment ,Antigens, CD19 ,Single Center ,Immunotherapy, Adoptive ,Gastroenterology ,Immune Reconstitution ,Internal medicine ,medicine ,Humans ,Pneumocystis jirovecii ,B-cell lymphoma ,Biological Products ,biology ,business.industry ,Hazard ratio ,Hematology ,Immunotherapy ,medicine.disease ,biology.organism_classification ,Lymphoma ,Platelet transfusion ,biology.protein ,Lymphoma, Large B-Cell, Diffuse ,Antibody ,business - Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 has significantly improved outcomes in the treatment of refractory or relapsed large B-cell lymphoma (LBCL). We evaluated the long-term course of hematologic recovery, immune reconstitution, and infectious complications in 41 patients with LBCL treated with axicabtagene ciloleucel (axi-cel) at a single center. Grade 3+ cytopenias occurred in 97.6% of patients within the first 28 days postinfusion, with most resolved by 6 months. Overall, 63.4% of patients received a red blood cell transfusion, 34.1% of patients received a platelet transfusion, 36.6% of patients received IV immunoglobulin, and 51.2% of patients received growth factor (granulocyte colony-stimulating factor) injections beyond the first 28 days postinfusion. Only 40% of patients had recovered detectable CD19+ B cells by 1 year, and 50% of patients had a CD4+ T-cell count
- Published
- 2021
20. Belumosudil Combination Therapy in Refractory Chronic Graft-Versus-Host Disease
- Author
-
Michelle Chin, Judith A. Shizuru, Lori Muffly, Parveen Shiraz, Laura J. Johnston, Robert Lowsky, Andrew R. Rezvani, Matthew J. Frank, Sushma Bharadwaj, Wen-Kai Weng, Robert S. Negrin, David B. Miklos, and Sally Arai
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
21. Orca-Q Demonstrates Favorable GvHD-and-Relapse-Free Survival in Haploidentical Transplants without Post-Transplant Cyclophosphamide
- Author
-
Amandeep Salhotra, Samer A Srour, Rasmus T Hoeg, Ayman Saad, Everett H Meyer, Anna Pavlova, Edmund K. Waller, Mwe Mwe Chao, J Scott McClellan, Nathaniel B Fernhoff, Robert Lowsky, and Mehrdad Abedi
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
22. Nonmyeloablative allogeneic transplantation achieves clinical and molecular remission in cutaneous T-cell lymphoma
- Author
-
Andrew R. Rezvani, Lori Muffly, Laura Johnston, Wen-Kai Weng, Youn H. Kim, Robert Lowsky, Judith A. Shizuru, Sally Arai, Robert S. Negrin, Richard T. Hoppe, Michael S. Khodadoust, David B. Miklos, Erica Wang, Everett Meyer, Timothy Almazan, Lynn Million, and Shufeng Li
- Subjects
medicine.medical_specialty ,Mycosis fungoides ,Skin Neoplasms ,Transplantation Conditioning ,Allogeneic transplantation ,Clinical Trials and Observations ,business.industry ,Incidence (epidemiology) ,Cutaneous T-cell lymphoma ,Hematology ,medicine.disease ,Minimal residual disease ,Gastroenterology ,Tacrolimus ,Lymphoma, T-Cell, Cutaneous ,Regimen ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Cumulative incidence ,Neoplasm Recurrence, Local ,business ,Aged - Abstract
The majority of patients with refractory, advanced-stage mycosis fungoides (MF) or Sézary syndrome (SS) have a life expectancy of
- Published
- 2020
23. Outcomes with autologous stem cell transplant vs. non-transplant therapy in patients 70 years and older with multiple myeloma
- Author
-
Surbhi Sidana, Christopher Lemieux, Robert S. Negrin, Matthew J. Frank, Sally Arai, Andrew R. Rezvani, Robert Lowsky, Wen-Kai Weng, Lori Muffly, Judith A. Shizuru, David J. Iberri, Laura Johnston, Michaela Liedtke, Parveen Shiraz, Everett Meyer, David B. Miklos, and Juliana Craig
- Subjects
medicine.medical_specialty ,Transplantation Conditioning ,Multivariate analysis ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Transplantation, Autologous ,Disease-Free Survival ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Multiple myeloma ,Aged ,Retrospective Studies ,Transplantation ,Performance status ,business.industry ,Hematopoietic Stem Cell Transplantation ,Retrospective cohort study ,Hematology ,medicine.disease ,Hematologic Response ,Treatment Outcome ,030220 oncology & carcinogenesis ,Cohort ,Multiple Myeloma ,business ,Stem Cell Transplantation ,030215 immunology - Abstract
We evaluated 79 patients with multiple myeloma (MM) ≥70 years referred to our blood and marrow transplant clinic, within 1 year of diagnosis from 2010 to 2019, for consideration of autologous stem cell transplant (ASCT). Thirty-eight (48%) of 79 patients underwent ASCT. ASCT was not pursued in 41 (52%) patients due to: patient or physician preference in 80% (n = 33) or ineligibility in 20% (n = 8). Baseline characteristics of patients in the two groups were similar. Median PFS from treatment start amongst patients undergoing ASCT (n = 38) vs. not (n = 41) was 41 months vs. 33 months, p = 0.03. There was no difference in OS, with estimated 5-year OS of 73% vs. 83%, respectively (p = 0.86). Day +100 transplant-related mortality (TRM) was 0%. ASCT was an independent favorable prognostic factor for PFS in multivariate analysis, after accounting for HCT-CI score, performance status, hematologic response, and maintenance. Finally, patients ≥70 years undergoing ASCT had similar PFS compared to a contemporaneous institutional cohort of patients
- Published
- 2020
24. Allogeneic Hematopoietic Cell Transplantation for Adult Acute Lymphoblastic Leukemia in the Modern Era
- Author
-
Emily C. Liang, Juliana Craig, Stefan Torelli, Kristen Cunanan, Maria Iglesias, Sally Arai, Matthew J. Frank, Laura Johnston, Robert Lowsky, Everett H. Meyer, David B. Miklos, Robert Negrin, Andrew Rezvani, Parveen Shiraz, Judith Shizuru, Surbhi Sidana, Wen-Kai Weng, Sushma Bharadwaj, and Lori Muffly
- Subjects
Adult ,Transplantation ,Neoplasm, Residual ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Tissue Donors ,Acute Disease ,Molecular Medicine ,Immunology and Allergy ,Humans ,Transplantation, Homologous - Abstract
Allogeneic hematopoietic cell transplantation (HCT) remains an important treatment for adults with acute lymphoblastic leukemia (ALL). We hypothesized that advances in ALL and transplantation have resulted in improved HCT outcomes in recent years. In this study, we evaluated the characteristics and outcomes of adult ALL patients undergoing allogeneic HCT over the last decade. Patients with ALL aged 18 years and older who underwent allogeneic HCT at Stanford University between 2008 and 2019 were included in this study. Patients were divided into 2 eras based on year of HCT: 2008 to 2013 (earlier era) and 2014 to 2019 (later era). A total of 285 patients were included: 119 patients underwent HCT in the earlier era and 166 in the later era. Patients who underwent transplantation in the later era were more likely to be Hispanic (38% versus 21%) and to have an HCT-comorbidity index ≥3 (31% versus 18%). Donor source for HCT also differed with an increase in the use of HLA-mismatched donor sources (38% versus 24%), notably umbilical cord blood in the later era (16% versus 0%). Patients in the later era were less likely to undergo transplantation with active disease (4% versus 16%); pre-HCT rates of measurable residual disease were similar across the eras (38% versus 40%). In unadjusted analyses, overall survival (OS) improved across eras, with 2-year estimates for the later and earlier eras of 73% (95% confidence interval [CI], 66%-80%) versus 55% (95% CI, 46%-64%), respectively. Multivariable analysis confirmed the association between later era and OS (hazard ratio = 0.52, 95% CI, 0.34-0.78). Finally, among patients relapsing after HCT (25% in later era and 33% in earlier era), the use of novel immunotherapies increased in the later era (44% versus 3%), as did the median OS after post-HCT relapse (16 months versus 8 months, P.001). OS after HCT for adult ALL has improved in recent years. This is due, in part, to a significant improvement in the ability to effectively salvage adults with ALL relapsing after HCT.
- Published
- 2022
25. Orca-T, a High-Precision Cell Therapy, for the Treatment of Hematologic Malignancies in Patients with 7/8 Mismatch Donors
- Author
-
Anna Pavlova, Robert Lowsky, Lori Muffly, Nathaniel B. Fernhoff, James Scott McClellan, Robert S. Negrin, and Everett H. Meyer
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
26. Analysis of Bendamustine Lymphodepletion, CD19 CART Expansion, Safety and Efficacy in Patients with Rel/Ref Non-Hodgkin Lymphoma
- Author
-
Sushma Bharadwaj, Mark P. Hamilton, Bita Sahaf, John Tamaresis, Theresa M. Latchford, Sally Arai, Laura Johnston, Robert Lowsky, Robert S. Negrin, Andrew R. Rezvani, Judith A. Shizuru, Everett H. Meyer, Parveen Shiraz, Surbhi Sidana, Melody Smith, Wen-Kai Weng, Lori Muffly, Crystal Mackall, Matthew J. Frank, David B. Miklos, and Saurabh Dahiya
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
27. Impact of Conditioning Regimen and Donor Type on Outcomes of Allogeneic Stem Cell Transplant for Myelofibrosis – a Single Institutional Experience
- Author
-
Judith A. Shizuru, Parveen Shiraz, Lori Muffly, Robert S. Negrin, Waqas Jehangir, Everett Meyer, Andrew R. Rezvani, David B. Miklos, Wen-Kai Weng, Surbhi Sidana, Sally Arai, Laura Johnston, Matthew J. Frank, and Robert Lowsky
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Conditioning regimen ,Internal medicine ,medicine ,Molecular Medicine ,Immunology and Allergy ,Stem cell ,business ,Myelofibrosis - Published
- 2021
28. Real-World Experience of Cryopreserved Allogeneic Hematopoietic Grafts during the COVID-19 Pandemic: A Single-Center Report
- Author
-
Andriyana K. Bankova, Joseph Caveney, Bin Yao, Teresa L. Ramos, Jan Bögeholz, Kartoosh Heydari, Nery Diaz, Marin L. Jackson, Robert Lowsky, Janice (Wes) Brown, Laura Johnston, Andrew R. Rezvani, Matthew J. Frank, Lori Muffly, Wen-Kai Weng, Surbhi Sidana, Robert S. Negrin, David B. Miklos, Parveen Shiraz, Everett H. Meyer, Judith A. Shizuru, Sally Arai, University of Zurich, and Arai, Sally
- Subjects
Cryopreservation ,Transplantation ,2747 Transplantation ,2720 Hematology ,Hematopoietic Stem Cell Transplantation ,COVID-19 ,Engraftment Failure ,Reduced intensity conditioning ,610 Medicine & health ,Cryopreserved allografts ,2700 General Medicine ,Cell Biology ,Hematology ,Article ,1307 Cell Biology ,Graft composition ,1313 Molecular Medicine ,10032 Clinic for Oncology and Hematology ,2723 Immunology and Allergy ,Molecular Medicine ,Immunology and Allergy ,Humans ,Neoplasm Recurrence, Local ,Pandemics ,Retrospective Studies - Abstract
Background As a result of the COVID-19 widespread pandemic, cryopreservation of allogeneic donor apheresis products was implemented to mitigate the challenges of donor availability and product transport. Although logistically beneficial, the impact of cryopreservation on clinical outcomes and graft composition remains unclear. Objectives To compare the outcomes and graft composition with cryopreserved versus fresh allografts in the setting of allogeneic hematopoietic cell transplantation (allo-HCT). Study design We retrospectively analyzed the clinical outcomes of 30 consecutive patients who received cryopreserved allografts between March and August 2020 as compared to 60 consecutive patients who received fresh allografts prior to the COVID-19 pandemic. Primary endpoints were hematopoietic engraftment, graft failure (GF) and secondary outcomes were overall survival (OS), relapse free survival (RFS) and non-relapse mortality (NRM). In addition, extended immunophenotype analysis was performed on cryopreserved versus prospectively collected fresh apheresis samples. Results Compared to fresh allografts, both neutrophil and platelet recovery were delayed in recipients of cryopreserved reduced intensity conditioning (RIC) allo-HCT with median times to engraftment of 24 days vs 18 days (P = .01) and 27 days vs 18 days (P = .069), respectively. We observed primary GF in 4 of 30 patients in the cryopreserved cohort (13.3%) vs only one of 60 patients (1.7 %) in the fresh cohort (P = .03). Cryopreserved RIC allo-HCT was associated with significantly lower median total, myeloid and T-cell donor chimerism at 1 month. OS and RFS were inferior for cryograft recipients with hazard ratio [HR (95%Cl)]: 2.16 (1.00, 4.67) and 1.90 (0.95, 3.79), respectively. Using an extended immunophenotype analysis we compared 14 samples from the cryopreserved cohort to 6 prospectively collected fresh apheresis donor samples. These analyses showed both decrease in total cell viability and significantly reduced absolute numbers of NK cells (CD3−CD56+) in the cryopreserved apheresis samples. Conclusion In this single institution study we note delayed engraftment and a trend toward clinical inferiority of cryopreserved vs fresh allografts. Further evaluation of the use of cryopreserved allografts and their impact on clinical and laboratory outcomes is warranted.
- Published
- 2021
29. Allogeneic transplantation after PD-1 blockade for classic Hodgkin lymphoma
- Author
-
Didier Blaise, Tycel Phillips, Jason T. Romancik, Roch Houot, Robert Lowsky, Martina Sollini, Jean Marc Schiano De Colella, Amer Beitinjaneh, Gunjan L. Shah, Lazaros J. Lekakis, Stephen D. Smith, Paolo Corradini, Mohamad Mohty, Maryam Rahimian, Luca Castagna, Taiga Nishihori, Asad Bashey, Talha Badar, Reid W. Merryman, Mehdi Hamadani, Carmelo Carlo-Stella, Dipenkumar Modi, Sally Arai, Kamal Bouabdallah, Valter Torri, Joseph P. McGuirk, Guillaume Manson, Anna Guidetti, Yi-Bin Chen, Hatcher J. Ballard, Julio C. Chavez, Pier Luigi Zinzani, Tatyana Feldman, Sunita Nathan, Anurag K. Singh, Massimo Magagnoli, Marie Pierre Moles-Moreau, Beatrice Casadei, Anthony Serritella, Michael Byrne, Radhakrishnan Ramchandren, Miguel-Angel Perales, Chiara De Philippis, Samantha Jaglowski, Justin Kline, Remy Dulery, Laura Giordano, Alex F. Herrera, Jonathon B. Cohen, Philippe Armand, Armando Santoro, Aspasia Stamatoulas, Stephen M. Ansell, Michael A. Spinner, Lori Dahncke, Corentin Orvain, Chloé Spilleboudt, Geoffrey Shouse, Robin Joyce, Vincent T. Ho, Matthew J. Frigault, Ryan C. Lynch, Uttam Rao, Jakub Svoboda, David A. Bond, Yago Nieto, Dana-Farber Cancer Institute [Boston], Humanitas Clinical and Research Center [Rozzano, Milan, Italy], Università degli Studi di Milano = University of Milan (UNIMI), University of Bologna/Università di Bologna, Ohio State University [Columbus] (OSU), Stanford University, Memorial Sloane Kettering Cancer Center [New York], Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), Institut Jules Bordet [Bruxelles], Faculté de Médecine [Bruxelles] (ULB), Université libre de Bruxelles (ULB)-Université libre de Bruxelles (ULB), CHU Saint-Antoine [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Centre de Lutte Contre le Cancer Henri Becquerel Normandie Rouen (CLCC Henri Becquerel), CHU Pontchaillou [Rennes], Microenvironment, Cell Differentiation, Immunology and Cancer (MICMAC), Université de Rennes (UR)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Centre Hospitalier Universitaire d'Angers (CHU Angers), PRES Université Nantes Angers Le Mans (UNAM), CHU Bordeaux [Bordeaux], Humanitas University [Milan] (Hunimed), P01 CA23766, U.S. Department of Health & Human Services | National Institutes of Health, U.S. Department of Health & Human Services | National Institutes of Health, 20575, Associazione Italiana per la Ricerca sul Cancro, Università degli Studi di Milano [Milano] (UNIMI), University of Bologna, Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Merryman R.W., Castagna L., Giordano L., Ho V.T., Corradini P., Guidetti A., Casadei B., Bond D.A., Jaglowski S., Spinner M.A., Arai S., Lowsky R., Shah G.L., Perales M.-A., De Colella J.M.S., Blaise D., Herrera A.F., Shouse G., Spilleboudt C., Ansell S.M., Nieto Y., Badar T., Hamadani M., Feldman T.A., Dahncke L., Singh A.K., McGuirk J.P., Nishihori T., Chavez J., Serritella A.V., Kline J., Mohty M., Dulery R., Stamatoulas A., Houot R., Manson G., Moles-Moreau M.-P., Orvain C., Bouabdallah K., Modi D., Ramchandren R., Lekakis L., Beitinjaneh A., Frigault M.J., Chen Y.-B., Lynch R.C., Smith S.D., Rao U., Byrne M., Romancik J.T., Cohen J.B., Nathan S., Phillips T., Joyce R.M., Rahimian M., Bashey A., Ballard H.J., Svoboda J., Torri V., Sollini M., De Philippis C., Magagnoli M., Santoro A., Armand P., Zinzani P.L., and Carlo-Stella C.
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,allogenic stem cell transplantation, PD-1 blockade, Hodgkin lymhpoma ,[SDV]Life Sciences [q-bio] ,Programmed Cell Death 1 Receptor ,Salvage therapy ,0302 clinical medicine ,Young adult ,Immune Checkpoint Inhibitors ,ComputingMilieux_MISCELLANEOUS ,Hematopoietic Stem Cell Transplantation ,Hematology ,Middle Aged ,Prognosis ,Hodgkin Disease ,3. Good health ,Survival Rate ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Adult ,medicine.medical_specialty ,Allogeneic transplantation ,Cyclophosphamide ,Adolescent ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,Young Adult ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Survival rate ,Aged ,Retrospective Studies ,Salvage Therapy ,business.industry ,Retrospective cohort study ,[SDV.IMM.IMM]Life Sciences [q-bio]/Immunology/Immunotherapy ,Blockade ,Transplantation ,030104 developmental biology ,Drug Resistance, Neoplasm ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
International audience; Anti-PD-1 monoclonal antibodies yield high response rates in patients with relapsed/refractory classic Hodgkin lymphoma (cHL), but most patients will eventually progress. Allogeneic hematopoietic cell transplantation (alloHCT) after PD-1 blockade may be associated with increased toxicity, raising challenging questions about the role, timing, and optimal method of transplantation in this setting. To address these questions, we assembled a retrospective cohort of 209 cHL patients who underwent alloHCT after PD-1 blockade. With a median follow-up among survivors of 24 months, the 2-year cumulative incidences (CIs) of non-relapse mortality and relapse were 14 and 18%, respectively; the 2-year graft-versus-host disease (GVHD) and relapse-free survival (GRFS), progression-free survival (PFS), and overall survival were 47%, 69%, and 82%, respectively. The 180-day CI of grade 3-4 acute GVHD was 15%, while the 2-year CI of chronic GVHD was 34%. In multivariable analyses, a longer interval from PD-1 to alloHCT was associated with less frequent severe acute GVHD, while additional treatment between PD-1 and alloHCT was associated with a higher risk of relapse. Notably, post-transplant cyclophosphamide (PTCy)-based GVHD prophylaxis was associated with significant improvements in PFS and GRFS. While awaiting prospective clinical trials, PTCy-based GVHD prophylaxis may be considered the optimal transplantation strategy for this patient population.
- Published
- 2021
30. 215.9: Achievement of Persistent Mixed Chimerism in Recipients of Matched and Mismatched Living Donor Kidney Transplants in a Tolerance Induction Protocol
- Author
-
John Scandling, Stephan Busque, Robert Lowsky, Thomas Pham, Richard Hoppe, Kent Jensen, Asha Shori, Hsin-Hsu Wu, Rahul Pawar, Edgar Engleman, Everett Meyer, and Sam Strober
- Subjects
Transplantation - Published
- 2022
31. High Resolution KIR in Non-Myeloablative Conditioning Regimen for Hematologic Neoplasms
- Author
-
Livia Caroline Barbosa Mariano, Gonzalo Martin, Leonardo Maldaner Amorim, Danillo G Augusto, Paul J Norman, Jill A Hollenbach, Robert Lowsky, Vanderson Rocha, and Marcelo Fernandez-Vina
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
32. Orca-T Demonstrates Encouraging Overall Survival, Gvhd Reduction, and Tolerability in Patients with Hematologic Malignancies
- Author
-
Anna Moroz, Rasmus Hoeg, Arpita Gandhi, Lori Muffly, Parveen Shiraz, Caspian Oliai, Rohtesh S. Mehta, Samer A Srour, Joseph P. McGuirk, Edmund K. Waller, Sally Arai, Laura Johnston, Robert Lowsky, Andrew R. Rezvani, Wen-Kai Weng, David B. Miklos, Matthew J. Frank, John Tamaresis, Vaibhav Agrawal, Nathaniel Fernhoff, Gerhard Bauer, Amy Putnam, James Scott McClellan, Bronwen E. Shaw, Mehrdad Abedi, Robert S. Negrin, and Everett H. Meyer
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
33. Outcomes after Delayed and Second Autologous Stem Cell Transplant in Patients with Relapsed Multiple Myeloma
- Author
-
Robert S. Negrin, Wen-Kai Weng, Sally Arai, Lori Muffly, Matthew J. Frank, David B. Miklos, Andrew R. Rezvani, Laura Johnston, Robert Lowsky, Judith A. Shizuru, Michaela Liedtke, Everett Meyer, Parveen Shiraz, David J. Iberri, Surbhi Sidana, Christopher Lemieux, and Juliana Craig
- Subjects
medicine.medical_specialty ,Transplantation, Autologous ,Article ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Maintenance therapy ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,In patient ,Stage (cooking) ,Multiple myeloma ,Retrospective Studies ,Salvage Therapy ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Surgery ,Clinical trial ,Prior Therapy ,030220 oncology & carcinogenesis ,Stem cell ,Neoplasm Recurrence, Local ,business ,Multiple Myeloma ,030215 immunology - Abstract
We evaluated the outcomes of 168 patients undergoing delayed or second autologous stem cell transplant (ASCT) for relapsed multiple myeloma (MM) from 2010 to 2019. Overall, 21% (n = 35) patients had received a prior transplant and 69% (n = 116) underwent transplant at first relapse. Overall, 27% patients had high-risk cytogenetics and 15% had ISS stage III disease. Stem cell collection was performed after relapse in 72% and 35% of patients received maintenance therapy. Median PFS from salvage treatment and transplant were 28 and 19 months, respectively. Median OS from salvage treatment and transplant was 69 and 55 months. Multivariate analysis revealed that ASCT in first relapse was associated with superior PFS (HR 0.63, p = 0.03) and OS (HR 0.59, p = 0.04) compared to later lines of therapy. In addition, PFS of ≥36 months with prior therapy was associated with improved PFS (HR 0.62, p = 0.04) and OS (HR 0.41, p = 0.01). Ninety-five patients underwent delayed transplant at first relapse, median PFS and OS from start of therapy was 30 and 69 months, and median OS from diagnosis was 106 months. These data may serve as a guide when counseling patients undergoing ASCT for relapsed MM and provide a benchmark in designing clinical trials of transplantation/comparative treatments for relapsed MM.
- Published
- 2021
34. Combined kidney and hematopoeitic cell transplantation to induce mixed chimerism and tolerance
- Author
-
Robert Lowsky and Samuel Strober
- Subjects
Transplantation Conditioning ,medicine.medical_treatment ,Human leukocyte antigen ,Chimerism ,Article ,Immune tolerance ,03 medical and health sciences ,0302 clinical medicine ,Cell transplantation ,Humans ,Medicine ,Transplantation ,Kidney ,Mixed chimerism ,business.industry ,Haplotype ,Hematopoietic Stem Cell Transplantation ,Hematology ,Kidney Transplantation ,Immunosuppressive drug ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,business ,030215 immunology - Abstract
Based on preclinical studies, combined kidney and hematopoietic cell transplantation was performed on fully HLA matched and haplotype matched patients at the Stanford University Medical Center. The object of the studies was to induce mixed chimerism, immune tolerance, and complete immunosuppressive drug withdrawal. Tolerance, persistent mixed chimerism, and complete withdrawal was achieved in the majority of fully matched patients. Persistent mixed chimerism and partial withdrawal has been achieved in the haplotype matched patients at present.
- Published
- 2019
35. Autologous Hematopoietic Cell Transplantation for Treatment-Refractory Relapsing Multiple Sclerosis: Position Statement from the American Society for Blood and Marrow Transplantation
- Author
-
Robert Lowsky, Paolo A. Muraro, Richard A. Nash, Laura E. Baldassari, Stefanie Sarantopoulos, Mark S. Freedman, James Bowen, Marcelo C. Pasquini, Paul A. Carpenter, Keith M. Sullivan, Christopher Bredeson, Jan Storek, Bipin N. Savani, John R. Corboy, Linda M. Griffith, Harold L. Atkins, Jeffrey A. Cohen, George E. Georges, Navneet S. Majhail, and National Institutes of Health
- Subjects
Canada ,medicine.medical_specialty ,Coverage ,medicine.medical_treatment ,Immunology ,indication ,Salvage therapy ,IMMUNOABLATION ,PROGRESSION ,Stem cells ,Hematopoietic stem cell transplantation ,multiple sclerosis ,Transplantation, Autologous ,ECTRIMS/EAN GUIDELINE ,03 medical and health sciences ,0302 clinical medicine ,LONG-TERM OUTCOMES ,Autologous hematopoietic cell transplantation ,PEOPLE ,Humans ,Medicine ,Intensive care medicine ,Adverse effect ,Societies, Medical ,Patient Care Team ,Salvage Therapy ,Transplantation ,Science & Technology ,business.industry ,DISABILITY ,Multiple sclerosis ,Hematopoietic Stem Cell Transplantation ,1103 Clinical Sciences ,Retrospective cohort study ,ADULTS ,Hematology ,medicine.disease ,United States ,Clinical trial ,Treatment Outcome ,Systematic review ,030220 oncology & carcinogenesis ,DISEASE-MODIFYING THERAPIES ,DOSE IMMUNOSUPPRESSIVE THERAPY ,business ,Life Sciences & Biomedicine ,PHARMACOLOGICAL-TREATMENT ,030215 immunology - Abstract
Multiple sclerosis (MS) is a chronic, disabling, immune-mediated, demyelinating and degenerative disease of the central nervous system. Approved disease-modifying therapies may be incompletely effective in some patients with highly active relapsing disease and high risk of disability. The use of immunoablative or myeloablative therapy followed by autologous hematopoietic cell transplantation (AHCT) has been investigated in retrospective studies, clinical trials, and meta-analyses/systematic reviews as an approach to address this unmet clinical need. On behalf of the American Society for Blood and Bone Marrow Transplantation (ASBMT), a panel of experts in AHCT and MS convened to review available evidence and make recommendations on MS as an indication for AHCT. A review of recent literature identified 8 retrospective studies, 8 clinical trials, and 3 meta-analyses/systematic reviews. In aggregate, these studies indicate that AHCT is an efficacious and safe treatment for active relapsing forms of MS to prevent clinical relapse, magnetic resonance imaging-detectable lesion activity, and worsening disability and to reverse disability without unexpected adverse events. Based on the available evidence, the ASBMT recommends that treatment-refractory relapsing MS with high risk of future disability be considered a "standard of care, clinical evidence available" indication for AHCT. Collaboration of neurologists with expertise in treating MS and transplantation physicians with experience performing AHCT for autoimmune disease is crucial for ensuring appropriate patient selection and optimizing transplantation procedures to improve patient outcomes. Transplantation centers in the United States and Canada are strongly encouraged to report baseline and outcomes data on patients receiving AHCT for multiple sclerosis to the Center for International Blood and Marrow Transplant Research.
- Published
- 2019
36. Incidence and risk factors associated with bleeding and thrombosis following chimeric antigen receptor T-cell therapy
- Author
-
Judith A. Shizuru, Jay Y. Spiegel, Lori Muffly, Matthew J. Frank, Surbhi Sidana, Wen-Kai Weng, John S. Tamaresis, Robert S. Negrin, Sally Arai, Andrew Johnsrud, Juliana Craig, James L. Zehnder, Robert Lowsky, Andrew R. Rezvani, David B. Miklos, Theresa Latchford, John H. Baird, Laura Johnston, Everett Meyer, Parveen Shiraz, and Crystal L. Mackall
- Subjects
medicine.medical_specialty ,Clinical Trials and Observations ,Cell- and Tissue-Based Therapy ,Fibrinogen ,Gastroenterology ,Risk Factors ,Internal medicine ,medicine ,Humans ,Platelet ,Retrospective Studies ,Univariate analysis ,Receptors, Chimeric Antigen ,business.industry ,Genitourinary system ,Incidence (epidemiology) ,Incidence ,Thrombosis ,Hematology ,medicine.disease ,Lymphoma ,Chimeric Antigen Receptor T-Cell Therapy ,business ,medicine.drug - Abstract
Key Points Clinically significant bleeding occurred in 9% of patients after CAR T therapy and was associated with features of systemic coagulopathy.Low baseline platelets and possibly high-grade ICANS are risk factors for bleeding and require close monitoring for bleeding up to 1 month., Visual Abstract, Bleeding and thrombotic events are an emerging toxicity associated with chimeric antigen receptor (CAR) therapies. To determine their incidence, we retrospectively analyzed consecutive adult patients (N = 127) with large B-cell lymphoma (LBCL) or B-cell acute lymphoblastic leukemia (B-ALL) treated from 2017 through 2020 with axicabtagene ciloleucel (axi-cel; n = 89) or a bispecific CD19/CD22 CAR (n = 38). Twelve (9.4%) and 8 (6.3%) patients developed bleeding and thrombosis within the first 3 months, respectively. In the axi-cel subgroup, these occurred in 11.2% and 6.7%, respectively. Bleeding occurred between days 8 and 30 (median, 17.5) and thrombosis between days 2 and 91 (median, 29). Bleeding sites included genitourinary, soft tissue, intracranial, gastrointestinal, and pulmonary and were associated with features of consumptive coagulopathy. On univariate analysis, patients with bleeding were older, had lower baseline platelets (86 × 103/μL vs 178 × 103/μL; P < .01), lower platelet and fibrinogen nadirs , and elevated lactate dehydrogenase. Immune effector cell (IEC)–associated neurotoxicity syndrome (ICANS) grade ≥3 was associated with increased bleeding (50% vs 15%; P = .01), thrombosis (50% vs 16%; P = .04), prothrombin time prolongation, hypofibrinogenemia, and elevated D-dimer. Low pretreatment platelet counts were associated with bleeding in a multivariate logistic regression model. Patients with thrombocytopenia or severe ICANS are at increased risk of bleeding and should be closely monitored, particularly within the first month after CAR therapy. Future studies in larger cohorts should assess risk factors for systemic coagulopathies in CAR T therapy, including their association with neurotoxicity.
- Published
- 2021
37. Stem Cell Mobilization in Multiple Myeloma: Comparing Safety and Efficacy of Cyclophosphamide +/- Plerixafor versus Granulocyte Colony-Stimulating Factor +/- Plerixafor in the Lenalidomide Era
- Author
-
Matthew J. Frank, David B. Miklos, Robert S. Negrin, Lori Muffly, Gary Goldstein, Andrew Johnsrud, Surbhi Sidana, Victoria Osgood, Laura Johnston, Robert Lowsky, Everett Meyer, Andrew R. Rezvani, Abdullah Ladha, Parveen Shiraz, Judith A. Shizuru, Sally Arai, and Wen-Kai Weng
- Subjects
Oncology ,medicine.medical_specialty ,Benzylamines ,Cyclophosphamide ,medicine.medical_treatment ,CD34 ,Antigens, CD34 ,Granulocyte ,Cyclams ,Article ,Heterocyclic Compounds ,Internal medicine ,Granulocyte Colony-Stimulating Factor ,Immunology and Allergy ,Medicine ,Humans ,Lenalidomide ,Multiple myeloma ,Retrospective Studies ,Transplantation ,business.industry ,Growth factor ,Plerixafor ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,medicine.disease ,Hematopoietic Stem Cell Mobilization ,Granulocyte colony-stimulating factor ,medicine.anatomical_structure ,Molecular Medicine ,business ,Multiple Myeloma ,medicine.drug - Abstract
Growth factor and chemotherapy-based stem cell mobilization strategies are commonly used to treat patients with multiple myeloma. We retrospectively compared 398 patients mobilized between 2017 and 2020 using either cyclophosphamide (4 g/m(2)) plus granulocyte colony-stimulating factor (G-CSF) or G-CSF alone, with on demand plerixafor (PXF) in both groups. Although total CD34(+) yield was higher after chemomobilization compared with G-CSF +/− PXF (median, 13.6 × 10(6)/kg versus 4.4 × 10(6)/kg; P < .01), achievement of ≥2 × 10(6) CD34(+) cells (95% versus 93.7%; P =.61) and rates of mobilization failure (5% versus 6.3%; P =.61) were similar. Fewer patients required PXF with chemomobilization (12.3% versus 49.5%; P < .01), and apheresis sessions were fewer (median, 1 [range, 1 to 4] versus 2 [range, 1 to 5]). The rate of complications, including neutropenic fever, emergency department visits, and hospitalizations, was higher after chemomobilization (30% versus 7.4%; P < .01). Previous use of ≤6 cycles of lenalidomide did not impair cell yield in either group. The median cost of mobilization was 17.4% lower in the G-CSF +/− PXF group (P = .01). Between group differences in time to engraftment were not clinically significant. Given similar rates of successful mobilization, similar engraftment time, and less toxicity and lower costs compared with chemomobilization, G-CSF with on-demand PXF may be preferable in myeloma patients with adequate disease control and limited lenalidomide exposure.
- Published
- 2021
38. Mgta-145 + Plerixafor Provides GCSF-Free Rapid and Reliable Hematopoietic Stem Cell Mobilization for Autologous Stem Cell Transplant in Patients with Multiple Myeloma: A Phase 2 Study
- Author
-
John S. Tamaresis, Andrew R. Rezvani, Kevin A. Goncalves, Judith A. Shizuru, Shaji Kumar, Matthew J. Frank, David B. Miklos, Ying Lu, Hitomi Hosoya, Laura Johnston, Douglas Girgenti, Anne Le, Sally Arai, Robert Lowsky, Andriyana K Bankova, Wen-Kai Weng, Everett Meyer, Parveen Shiraz, Lori Muffly, Surbhi Sidana, John C. Davis, and Veit Schmelmer
- Subjects
Transplantation ,business.industry ,Plerixafor ,Immunology ,Phases of clinical research ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,Stem cell ,business ,Multiple myeloma ,Hematopoietic Stem Cell Mobilization ,medicine.drug - Abstract
Background: MGTA-145, a CXCR2 agonist, has shown promising activity for hematopoietic stem cell (HSC) mobilization with plerixafor in pre-clinical models and healthy volunteers. It has the potential to become the first GCSF free regimen for HSC mobilization/apheresis in preparation for transplant, with fewer side effects, better patient experience and optimal resource utilization. Methods: We conducted a single center phase 2 trial of MGTA-145 + plerixafor for HSC mobilization in patients with multiple myeloma (MM), NCT04552743. Primary endpoint was collection of ≥2 x 10 6 CD34+ cells/kg in up to 2 days of mobilization/apheresis. Secondary endpoints were collection of 4 and 6 x 10 6 CD34+ cells/kg, safety and engraftment. Patients with MM, 18-70 years of age, within 1 year of starting treatment & CrCl > 30 ml/min were eligible. Patients received plerixafor 0.24 mg/kg (0.16 mg/kg if renal dysfunction) SQ, followed 2 hours later by MGTA-145 (0.03 mg/kg) IV over 3-10 minutes and apheresis within 30 minutes. Mobilization and collection were repeated for a second consecutive day if day 1 yield was < 6 x 10 6 CD34+ cells/kg. The study was open-label single arm trial of 15 patients. If 13 or more patients met primary endpoint, an expansion cohort of 10 patients was planned. The trial has 85% power at a 5% one-sided type I error rate. Our analysis is based on aggregated results from total cohort of 25 patients. Results: Median age was 62 years, 52% were female, 24% had ISS stage 3, 57% had high-risk FISH (primarily gain1q). Induction therapy was VRD in 68% (17), daratumumab VRD in 24% (6), CyBorD in 8% (2) patients. Median duration of induction was 4 months (3-6) and median lenalidomide exposure was 5 cycles (1-8), with > VGPR in 88% of patients. (Table 1) Plerixafor 0.24 mg/kg was given in 24 (96%) patients. Median pre-apheresis CD34 count was day 1, 24/uL (3-99) & day 2, 15/uL (5-46). Median total HSC cell yield (CD34+ cells/kg x 10 6) was 5.0 (1.1-16.2), day 1 yield was 3.4 (0.3-16.2) and day 2 yield was 1.9 (0.5-4.6) (Fig1). 88% (n=22) of patients met the primary endpoint of collecting sufficient HSCs in < 2 days of mobilization + apheresis to proceed to transplant, 68% (17) in 1 day (2 x 10 6 CD34+ cells/kg). 3 patients who did not meet primary endpoint successfully collected HSCs with standard GCSF + plerixafor dosing & 2-3 apheresis sessions. Secondary endpoints of 4 and 6 x 10 6 CD34+ cells/kg in < 2 days were met in 68% (17) and 40% (10) patients. MGTA-145+plerixafor was well tolerated. At least 1 treatment emergent AE (TEAE) with MGTA-145 was seen in 60% of patients (grade 1, n= 14, grade 2, n=1). Pain (all grade 1) was most common, seen in 44% (11), with 38% (9) patients experiencing acute onset transient bone pain with MGTA-145 (duration: 7 minutes, range: 3-28). Using the validated Brief Pain Inventory, 56% (14) patients self-reported pain with mobilization vs 40% (10) at baseline. 7/15 patients without baseline pain reported pain with mobilization. An increase for worst pain was seen on mobilization day 1, that returned to the baseline, but no difference for aggregate score of pain severity/interference (linear mixed effects model). At last follow-up, 18 patients have completed transplant with MGTA-145 mobilized graft, with melphalan 200 mg/m 2 in 15 (83%) patients. Median of 3.5 (2.2-8.1) x 10 6 CD34+ cells/kg were infused. All patients have engrafted. Median time to neutrophil engraftment of 12 days (range: 11-15) & platelet engraftment (platelets > 20,000, no transfusion in 7 days) of 17.5 days (15-33) are comparable to historical data (DiPersio et al. 2009). RBC transfusion was needed in 3 (17%) patients. 14 patients have day 100 follow-up, all with durable engraftment. MGTA-145 + plerixafor mobilized grafts had a favorable graft composition. We observed high enrichment for CD90+CD45RA- among CD34+ cells, a CD34 subset of long term engrafting HSCs (median: 36% of CD34+ cells, range 10-66%, N=25). 74% (17 of 23) grafts were minimal residual disease negative with next generation flow cytometry. Conclusions: This is the first study to evaluate the novel G-CSF-free regimen of MGTA-145 + plerixafor for HSC cell mobilization & collection for hematologic malignancies. The study cohort was representative of transplant eligible patients with MM, with 88% patients meeting the primary endpoint. The regimen was well tolerated. Patients achieved timely & durable engraftment. Our data support further development of this regimen for rapid HSC mobilization. Figure 1 Figure 1. Disclosures Sidana: Janssen: Consultancy, Research Funding; Allogene: Research Funding; Magenta Therapeutics: Consultancy, Research Funding; BMS: Consultancy. Kumar: Bluebird Bio: Consultancy; Adaptive: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Carsgen: Research Funding; Astra-Zeneca: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck: Research Funding; Oncopeptides: Consultancy; KITE: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Tenebio: Research Funding; Takeda: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Beigene: Consultancy; Antengene: Consultancy, Honoraria; Amgen: Consultancy, Research Funding; Roche-Genentech: Consultancy, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding; BMS: Consultancy, Research Funding; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Research Funding. Muffly: Pfizer, Amgen, Jazz, Medexus, Pfizer: Consultancy; Astellas, Jasper, Adaptive, Baxalta: Research Funding; Adaptive: Honoraria, Other: fees for non-CME/CE services: , Research Funding. Arai: Magenta Therapeutics: Research Funding. Meyer: Indee, Jura: Consultancy; Orca Biosystems: Research Funding; GigaImmune: Current holder of stock options in a privately-held company; Triursus Therapeutics: Current holder of stock options in a privately-held company. Rezvani: US Department of Justice: Consultancy; Kaleido: Other: One-time scientific advisory board; Nohla Therapeutics: Other: One-time scientific advisory board; Pharmacyclics-Abbvie: Research Funding. Weng: Kite Pharma: Research Funding. Frank: Kite-Gilead: Membership on an entity's Board of Directors or advisory committees; Allogene Therapeutics: Research Funding; Adaptive Biotechnologies: Research Funding. Shiraz: Kite Pharma-Gilead: Research Funding. Girgenti: Magenta Therapeutics: Current Employment. Goncalves: Magenta Therapeutics: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Schmelmer: Magenta Therapeutics: Current Employment, Current equity holder in publicly-traded company, Patents & Royalties. Davis: Magenta Therapeutics: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Shizuru: Jasper Therapeutics, Inc.: Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Other: Chair of scientific advisory board; Forty seven Inc: Other: Inventor on a patent licenses by Forty Seven. Forty seven was acquired by Gilead in 2020. Miklos: Pharmacyclics: Patents & Royalties; Kite, a Gilead Company, Amgen, Atara, Wugen, Celgene, Novartis, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Bioscience, Adicet, Pharmacyclics, Janssen, Takeda, Adaptive Biotechnologies and Miltenyi Biotechnologies: Consultancy; Pharmacyclics, Amgen, Kite, a Gilead Company, Novartis, Roche, Genentech, Becton Dickinson, Isoplexis, Miltenyi, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Biosciences, Adicet, Adaptive Biotechnologies: Research Funding; Adaptive Biotechnologies, Novartis, Juno/Celgene-BMS, Kite, a Gilead Company, Pharmacyclics-AbbVie, Janssen, Pharmacyclics, AlloGene, Precision Bioscience, Miltenyi Biotech, Adicet, Takeda: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2022
39. Development of immunosuppressive myeloid cells to induce tolerance in solid organ and hematopoietic cell transplant recipients
- Author
-
Thomas Hsin-Hsu Wu, Judith A. Shizuru, Robert Lowsky, Jeffrey Waters, Edgar G. Engleman, Robert S. Negrin, Pingping Zheng, Jeanette Baker, John D. Scandling, Rahul D. Pawar, Stephan Busque, Kent P. Jensen, Asha Shori, John S. Tamaresis, Samuel Strober, Holden T. Maecker, Suparna Dutt, Everett Meyer, David Hongo, Xuhuai Ji, Anirudh Saraswathula, and Philip W. Lavori
- Subjects
medicine.medical_specialty ,Clinical Trials as Topic ,Transplantation Conditioning ,Immunobiology and Immunotherapy ,business.industry ,medicine.medical_treatment ,Hematopoietic Stem Cell Transplantation ,Immunosuppression ,Hematology ,Hematopoietic stem cell transplantation ,Organ transplantation ,Transplant Recipients ,Immune tolerance ,Transplantation ,Mice ,Cytokine ,Immunology ,medicine ,Myeloid-derived Suppressor Cell ,Immune Tolerance ,Animals ,Humans ,Cytokine secretion ,Myeloid Cells ,business - Abstract
Replacement of failed organs followed by safe withdrawal of immunosuppressive drugs has long been the goal of organ transplantation. We studied changes in the balance of T cells and myeloid cells in the blood of HLA-matched and -mismatched patients given living donor kidney transplants followed by total lymphoid irradiation, anti-thymocyte globulin conditioning, and donor hematopoietic cell transplant to induce mixed chimerism and immune tolerance. The clinical trials were based on a conditioning regimen used to establish mixed chimerism and tolerance in mice. In preclinical murine studies, there was a profound depletion of T cells and an increase in immunosuppressive polymorphonuclear (pmn) myeloid-derived suppressor cells (MDSCs) in the spleen and blood following transplant. Selective depletion of pmn MDSCs in mice abrogated mixed chimerism and tolerance. In our clinical trials, patients given an analogous tolerance conditioning regimen developed similar changes, including profound depletion of T cells and a marked increase in MDSCs in blood posttransplant. Posttransplant pmn MDSCs transiently increased expression of lectin-type oxidized LDL receptor-1, a marker of immunosuppression, and production of the T-cell inhibitor arginase-1. These posttransplant pmn MDSCs suppressed the activation, proliferation, and inflammatory cytokine secretion of autologous T-cell receptor microbead-stimulated pretransplant T cells when cocultured in vitro. In conclusion, we elucidated changes in receptors and function of immunosuppressive myeloid cells in patients enrolled in the tolerance protocol that were nearly identical to those of MDSCs required for tolerance in mice. These trials were registered at www.clinicaltrials.gov as #NCT00319657 and #NCT01165762.
- Published
- 2020
40. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial
- Author
-
Steven A. Feldman, Kara L. Davis, John H. Baird, Robert Lowsky, Rachel C. Lynn, Magali Bazzano, Judith A. Shizuru, Matthew J. Frank, Surbhi Sidana, Maria Caterina Rotiroti, Maria Iglesias, Sean Mackay, Sally Arai, Bita Sahaf, Shabnum Patel, Nirali N. Shah, Laura Johnston, Jay Y. Spiegel, Jing Zhou, Juliana Craig, Robert S. Negrin, Robbie G. Majzner, Andrew R. Rezvani, Zach Ehlinger, Ilan R. Kirsch, Parveen Shiraz, Chelsea D. Mullins, Michael G. Ozawa, Nikolaos Gkitsas, Crystal L. Mackall, Terry J. Fry, Warren D. Reynolds, Yasodha Natkunam, Sneha Ramakrishna, Scott J. Bornheimer, Allison P. Jacob, Lori Muffly, Jean Oak, Haiying Qin, Katherine A. Kong, Wen-Kai Weng, Everett Meyer, Nasheed Hossain, John S. Tamaresis, Sheren F. Younes, David B. Miklos, Liora M. Schultz, Eric J Yang, and Harshini Chinnasamy
- Subjects
0301 basic medicine ,Oncology ,Adult ,medicine.medical_specialty ,Lymphoma, B-Cell ,Lymphoma ,medicine.medical_treatment ,Sialic Acid Binding Ig-like Lectin 2 ,Antigens, CD19 ,Cancer immunotherapy ,Immunotherapy, Adoptive ,General Biochemistry, Genetics and Molecular Biology ,CD19 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,immune system diseases ,Recurrence ,Phase I trials ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,B-cell lymphoma ,B cell ,Aged ,Acute lymphocytic leukaemia ,biology ,business.industry ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,Chimeric antigen receptor ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Disease Progression ,business ,Progressive disease - Abstract
Despite impressive progress, more than 50% of patients treated with CD19-targeting chimeric antigen receptor T cells (CAR19) experience progressive disease. Ten of 16 patients with large B cell lymphoma (LBCL) with progressive disease after CAR19 treatment had absent or low CD19. Lower surface CD19 density pretreatment was associated with progressive disease. To prevent relapse with CD19− or CD19lo disease, we tested a bispecific CAR targeting CD19 and/or CD22 (CD19-22.BB.z-CAR) in a phase I clinical trial (NCT03233854) of adults with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL) and LBCL. The primary end points were manufacturing feasibility and safety with a secondary efficacy end point. Primary end points were met; 97% of products met protocol-specified dose and no dose-limiting toxicities occurred during dose escalation. In B-ALL (n = 17), 100% of patients responded with 88% minimal residual disease-negative complete remission (CR); in LBCL (n = 21), 62% of patients responded with 29% CR. Relapses were CD19−/lo in 50% (5 out of 10) of patients with B-ALL and 29% (4 out of 14) of patients with LBCL but were not associated with CD22−/lo disease. CD19/22-CAR products demonstrated reduced cytokine production when stimulated with CD22 versus CD19. Our results further implicate antigen loss as a major cause of CAR T cell resistance, highlight the challenge of engineering multi-specific CAR T cells with equivalent potency across targets and identify cytokine production as an important quality indicator for CAR T cell potency., Bispecific CAR T cells targeting CD19 and CD22 exhibit clinical activity and low toxicity in patients with large B cell lymphoma and B cell acute lymphoblastic leukemia, with relapses associated with loss of CD19 but not CD22.
- Published
- 2020
41. Outcomes with Autologous or Allogeneic Stem Cell Transplantation in Patients with Plasma Cell Leukemia in the Era of Novel Agents
- Author
-
Everett Meyer, Robert Lowsky, Robert S. Negrin, Matthew J. Frank, Juliana Craig, Judith A. Shizuru, Wen-Kai Weng, Laura Johnston, Surbhi Sidana, Christopher Lemieux, David B. Miklos, Sally Arai, Lori Muffly, Parveen Shiraz, and Andrew R. Rezvani
- Subjects
medicine.medical_specialty ,Transplantation, Autologous ,Disease-Free Survival ,Article ,Leukemia, Plasma Cell ,03 medical and health sciences ,0302 clinical medicine ,Maintenance therapy ,medicine ,Humans ,Transplantation, Homologous ,Cause of death ,Retrospective Studies ,Plasma cell leukemia ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Retrospective cohort study ,Hematology ,Middle Aged ,medicine.disease ,Surgery ,Clinical trial ,Regimen ,Treatment Outcome ,Pharmaceutical Preparations ,030220 oncology & carcinogenesis ,Cohort ,business ,030215 immunology ,Stem Cell Transplantation - Abstract
Plasma cell leukemia (PCL) is a rare and very aggressive plasma cell disorder. The optimal treatment approach, including whether to pursue an autologous (auto) or allogeneic (allo) stem cell transplantation (SCT) is not clear, given the lack of clinical trial-based evidence. This single-center retrospective study describes the outcomes of 16 patients with PCL (n = 14 with primary PCL) who underwent either autoSCT (n = 9) or alloSCT (n = 7) for PCL in the era of novel agents, between 2007 and 2019. The median age of the cohort was 58 years. High-risk cytogenetics were found in 50% of the patients. All patients received a proteasome inhibitor and/or immunomodulatory drug-based regimen before transplantation. At the time of transplantation, 10 patients (62%) obtained at least a very good partial response (VGPR). The response after autoSCT (3 months) was at least a VGPR in 6 patients (67%; complete response [CR] in 5). All patients undergoing alloSCT achieved a CR at 3 months. Maintenance therapy was provided to 5 patients (56%) after autoSCT. The median progression-free survival after transplantation was 6 months in the autoSCT group, compared with 18 months in the alloSCT group (P = .09), and median overall survival (OS) after transplantation in the 2 groups was 19 months and 40 months, respectively (P = .41). The median OS from diagnosis was 27 months and 49 months, respectively (P = .50). Of the 11 deaths, 10 patients (91%) died of relapsed disease. AlloSCT was not observed to offer any significant survival advantage over autoSCT in PCL, in agreement with recent reports, and relapse remains the primary cause of death in these patients.
- Published
- 2020
42. Mixed chimerism and acceptance of kidney transplants after immunosuppressive drug withdrawal
- Author
-
Hsin-Hsu Wu, Kent P. Jensen, Thomas A. Pham, Philip W. Lavori, John D. Scandling, Marcelo A. Fernandez Viña, Everett Meyer, Jeffrey Waters, Stephan Busque, Kevin Sheehan, Asha Shori, Okmi Choi, Judith A. Shizuru, Robert Lowsky, Richard T. Hoppe, John S. Tamaresis, Samuel Strober, and Edgar G. Engleman
- Subjects
Adult ,Male ,0301 basic medicine ,Isoantigens ,T-Lymphocytes ,medicine.medical_treatment ,Naive B cell ,Human leukocyte antigen ,030230 surgery ,Chimerism ,Tacrolimus ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,B cell ,Kidney transplantation ,B-Lymphocytes ,business.industry ,Histocompatibility Testing ,Graft Survival ,General Medicine ,Middle Aged ,medicine.disease ,Kidney Transplantation ,Survival Analysis ,Tissue Donors ,Transplantation ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Immunosuppressive drug ,Haplotypes ,Withholding Treatment ,Immunology ,Female ,Lymphocyte Culture Test, Mixed ,business ,Immunosuppressive Agents - Abstract
Preclinical studies have shown that persistent mixed chimerism is linked to acceptance of organ allografts without immunosuppressive (IS) drugs. Mixed chimerism refers to continued mixing of donor and recipient hematopoietic cells in recipient tissues after transplantation of donor cells. To determine whether persistent mixed chimerism and tolerance can be established in patients undergoing living donor kidney transplantation, we infused allograft recipients with donor T cells and hematopoietic progenitors after posttransplant lymphoid irradiation. In 24 of 29 fully human leukocyte antigen (HLA)–matched patients who had persistent mixed chimerism for at least 6 months, complete IS drug withdrawal was achieved without subsequent evidence of rejection for at least 2 years. In 10 of 22 HLA haplotype–matched patients with persistent mixed chimerism for at least 12 months, reduction of IS drugs to tacrolimus monotherapy was achieved. Withdrawal of tacrolimus during the second year resulted in loss of detectable chimerism and subsequent rejection episodes, unless tacrolimus therapy was reinstituted. Posttransplant immune reconstitution of naïve B cells and B cell precursors was more rapid than the reconstitution of naïve T cells and thymic T cell precursors. Robust chimerism was observed only among naïve T and B cells but not among memory T cells. No evidence of rejection was observed in all surveillance graft biopsies obtained from mixed chimeric patients withdrawn from IS drugs, and none developed graft-versus-host disease. In conclusion, persistent mixed chimerism established in fully HLA- or haplotype-matched patients allowed for complete or partial IS drug withdrawal without rejection.
- Published
- 2020
43. Utilization of Backup Stem Cells for Stem Cell Boost and Second Transplant in Patients with Multiple Myeloma Undergoing Autologous Stem Cell Transplantation
- Author
-
Emily C. Liang, Lori Muffly, Parveen Shiraz, Judith A. Shizuru, Laura Johnston, Sally Arai, Wen-Kai Weng, Robert Lowsky, Andrew R. Rezvani, Everett H. Meyer, Matthew J. Frank, Robert S. Negrin, David B. Miklos, and Surbhi Sidana
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2021
44. Concordance of Next Generation Sequencing-Based Measurable Residual Disease between Peripheral Blood and Bone Marrow in Adults with Acute Lymphoblastic Leukemia Receiving Cellular Therapies
- Author
-
Robert S. Negrin, Robert Lowsky, Matthew J. Frank, Judith A. Shizuru, Everett Meyer, Andrew R. Rezvani, Laura Johnston, Parveen Shiraz, David B. Miklos, Wen-Kai Weng, Vandana Sundaram, Surbhi Sidana, Sally Arai, and Lori Muffly
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,business.industry ,Concordance ,Lymphoblastic Leukemia ,Cell Biology ,Hematology ,Disease ,Peripheral blood ,DNA sequencing ,medicine.anatomical_structure ,Internal medicine ,medicine ,Molecular Medicine ,Immunology and Allergy ,Bone marrow ,business - Published
- 2021
45. Bleeding and Thrombosis Are Associated with Endothelial Dysfunction in CAR-T Cell Therapy and Are Increased in Patients Experiencing Neurologic Toxicity
- Author
-
Matthew J. Frank, Andrew R. Rezvani, Robert Lowsky, Surbhi Sidana, Andrew Johnsrud, Laura Johnston, Sally Arai, Robert S. Negrin, David B. Miklos, Parveen Shiraz, Juliana Craig, Crystal L. Mackall, Wen-Kai Weng, James L. Zehnder, John H. Baird, Judith A. Shizuru, Everett Meyer, Theresa Latchford, Jay Y. Spiegel, and Lori Muffly
- Subjects
Transplantation ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Immunology ,Retrospective cohort study ,Cell Biology ,Hematology ,Thrombin time ,medicine.disease ,Biochemistry ,Thrombosis ,Lymphoma ,Cytokine release syndrome ,Internal medicine ,Cohort ,medicine ,Coagulopathy ,Molecular Medicine ,Immunology and Allergy ,Platelet ,business - Abstract
Background Treatment with chimeric antigen receptor (CAR) T cell therapies have shown dramatic, often durable responses for relapsed/refractory B-cell malignancies. However, it can be associated with significant side effects such as cytokine release syndrome (CRS), immune effector-cell associated neurotoxicity syndrome (ICANS) and life-threatening consumptive coagulopathies. The underlying pathobiology of such hemostatic defects and their distinct clinical sequelae remains obscure. This retrospective study aims at quantifying CAR T therapy associated bleeding and thrombotic complications and their association with CRS, ICANS, and laboratory derangements. Methods 130 adult patients with DLBCL or B-ALL treated between 2017-2020 with CD19 CAR-T therapy axicabtagene ciloleucel (N=90) or a bispecific CD 19/22 CAR construct utilizing 4-1BB costimulatory domains (N=40) were analyzed to determine dynamics of coagulation parameters and platelet counts as well as incidences of bleeding or thrombosis in the first three months after CAR T infusion. Events were included if graded ≥ 2 or if intervention was required. Platelet counts and coagulation parameters were collected prior to lymphodepletion (pre-LD), day 0, 3, 7, 14, 21, 28, 60 and 90. Results 12 (9.2%) and 8 (6.2%) patients developed bleeding and thrombotic complications in the first three months after CAR-T infusion, respectively. Events are characterized in Figure 1. All bleeding events occurred between days 0-30 (median 17.5, range 8-30), while thrombotic events occurred between days 2-91 (median day 29, range, 2-91). Two (1.5%) patients experienced both bleeding and thrombosis. Bleeding events coincided with the onset of thrombocytopenia and hypofibrinogenemia, and patients who bled had lower platelet (median 22.5 vs. 47 K/uL; p=0.03) and fibrinogen (median 151 vs. 351 ug/mL; p=0.007) nadirs in the first 30 days compared to those without bleeding. Temporally, the lowest median platelet nadir occurred at day 7 in patients with bleeding events vs. day 21 in patients without bleeding, while timing of fibrinogen nadirs were at day 21 in both. Patients with bleeding episodes were more likely to be older (median age: 70 vs. 60 yrs, p=0.03), have thrombocytopenia prior to lymphodepletion therapy (median 117.5 vs. 174.5 K/uL, p=0.01), and have elevated LDH (lymphoma subgroup; p=0.07). Other lab derangements in the first 30 days seen more frequently in patients with bleeding included prolonged thrombin time (TT) (21% vs. 6%; p=0.02), PT (16% vs. 5%; p=0.06), and elevated d-dimer (16% vs. 3%; p=0.01) indicative of a consumptive process. Thrombotic events were not significantly associated with elevated or peak d-dimer values (median 4.97 vs. 2.37 ug/mL, p=0.20). Interestingly, occurrence or severity of CRS was not associated with bleeding or thrombotic events, nor was it associated with marked derangements in coagulation abnormalities. However, higher grade ICANS (grade > 3) was associated with bleeding (42% vs. 15%; p=0.038), thrombosis (50% vs. 16%; p=0.03), and evidence of endothelial activation including PT prolongation (78% vs. 35%; p 13 (10%) patients received anticoagulation for prophylaxis or therapeutic indications that predated CAR T infusion. Four started anticoagulation secondarily for thrombotic events after CAR-T infusion, and one received tissue plasminogen activator (tPA) for an acute stroke. In this group, no patients developed bleeding complications from anticoagulation. Conclusion Both bleeding (9.2%), and thrombotic (6.2%) events are observed after CAR T cell therapy, with bleeding limited to the first month in our cohort. Notably, ICANS was uniquely associated with PT prolongation, hypofibrinogenemia, and increased fibrin degradation, in addition to both bleeding and thrombosis. These results suggest that a systemic coagulopathy coincides with high grade ICANS and whether these neurologic events truly represent sequelae of widespread vascular dysfunction warrants further investigation. Anticoagulation was safe in the patients whom it was indicated. Risk factors for bleeding and thrombotic complications should be studied prospectively to develop risk-assessment models and clinical guidelines for management of bleeding and thrombosis (including prophylaxis) during CAR T therapy. Disclosures Muffly: Adaptive: Research Funding; Servier: Research Funding; Amgen: Consultancy. Negrin:BioEclipse Therapeutics: Current equity holder in private company; Magenta Therapeutics: Consultancy, Current equity holder in publicly-traded company; KUUR Therapeutics: Consultancy; Biosource: Current equity holder in private company; Amgen: Consultancy; UpToDate: Honoraria. Shizuru:Jasper Therapeutics, Inc: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees. Meyer:Orca Bio: Research Funding. Shiraz:Kite, a Gilead Company: Research Funding; ORCA BioSystems: Research Funding. Rezvani:Pharmacyclics: Research Funding. Mackall:Apricity Health: Consultancy, Current equity holder in private company; NeoImmune Tech: Consultancy; Nektar Therapeutics: Consultancy; Allogene: Current equity holder in publicly-traded company; BMS: Consultancy; Lyell Immunopharma: Consultancy, Current equity holder in private company. Miklos:Adaptive Biotech: Consultancy, Other: Travel support, Research Funding; Kite-Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel support, Research Funding; Juno-Celgene-Bristol-Myers Squibb: Consultancy, Other: Travel support, Research Funding; Allogene Therapeutics Inc.: Research Funding; Novartis: Consultancy, Other: Travel support, Research Funding; Pharmacyclics: Consultancy, Other: Travel support, Patents & Royalties, Research Funding; Janssen: Consultancy, Other: Travel support; Miltenyi Biotec: Research Funding. Sidana:Janssen: Consultancy.
- Published
- 2021
46. Orca-T, a Precision Treg-Engineered Donor Product, in Myeloablative HLA-Matched Transplantation Prevents Acute Gvhd with Less Immunosuppression in an Early Multicenter Experience
- Author
-
Kartoosh Heydari, Robert Lowsky, Laura Johnston, Nathaniel Fernhoff, Hsin-Hsu Wu, Arpita Ghandi, Bryan J. Xie, Rahul D. Pawar, Rasmus T. Hoeg, James Scott McClellan, Caspian Oliai, Parveen Shiraz, Everett Meyer, Andrew R. Rezvani, Gerhard Bauer, Mehrdad Abedi, Judith A. Shizuru, Robert S. Negrin, David B. Miklos, Anna Moroz, Wen-Kai Weng, Sally Arai, Bronwen E. Shaw, Lori Muffly, and Joseph P. McGuirk
- Subjects
Transplantation ,business.industry ,medicine.medical_treatment ,Immunosuppression ,Cell Biology ,Hematology ,Human leukocyte antigen ,Product (mathematics) ,Immunology ,medicine ,Molecular Medicine ,Immunology and Allergy ,business - Published
- 2021
47. CD22-CAR T-Cell Therapy Mediates High Durable Remission Rates in Adults with Large B-Cell Lymphoma Who Have Relapsed after CD19-CAR T-Cell Therapy
- Author
-
Robert S. Negrin, Jean Oak, Kara L. Davis, Lori Muffly, Sheren F. Younes, Bita Sahaf, Juliana Craig, Maria Iglesias, Yasodha Natkunam, David B. Miklos, Shabnum Patel, Robert Lowsky, Sneha Ramakrishna, Surbhi Sidana, Wen-Kai Weng, Jay Y. Spiegel, Andrew R. Rezvani, Laura Johnston, Parveen Shiraz, Sally Arai, Emma Crawford, Zachary Ehlinger, Crystal L. Mackall, Steven A. Feldman, Emily Egeler, John H. Baird, Matthew J. Frank, Warren D. Reynolds, Liora M. Schultz, Harshini Chinnasamy, and Hrishikesh K. Srinagesh
- Subjects
biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,CD19 ,Cell therapy ,Cancer research ,medicine ,biology.protein ,CAR T-cell therapy ,B-cell lymphoma ,business ,CD22 CAR-T - Abstract
BACKGROUND: Patients with relapsed/refractory (R/R) large B-cell lymphoma (LBCL) after failure of CD19-directed CAR T-cell therapy (CAR19) have a dire prognosis, with an overall response rate (ORR) of 29% to conventional salvage therapies, and a median overall survival (OS) of 6 months. CD22 is expressed on the majority of B-cell malignancies. Autologous CAR T-cells targeting CD22 (CAR22) have yielded an ORR of 70-90% in pediatric patients with R/R B-cell acute lymphoblastic leukemia (B-ALL), including those who had previously failed CAR19 therapy. Based on these encouraging results, we evaluated CAR22 in adult patients with R/R LBCL, focusing on those with CAR19-refractory disease. METHODS: This ongoing single-institution phase I dose escalation clinical trial (NCT04088890) is evaluating a CAR construct incorporating the m971 CD22 single chain variable fragments and 41BB/CD3z endodomains integrated within autologous T-cells via lentiviral transduction. After lymphodepletion (LD) with fludarabine and cyclophosphamide, patients are infused with cryopreserved CAR T-cells after a 7- to 11-day closed manufacturing process utilizing the CliniMACS Prodigy device (Miltenyi). Primary objectives assess the ability to successfully manufacture CAR22 and safety. Secondary objectives include efficacy and durability of responses. RESULTS: Twenty-one patients with LBCL [n=12 at dose level 1 (DL1), 1x10 6 CAR+ cells/kg; n=9 at dose level 2 (DL2), 3x10 6 CAR+ cells/kg] have been enrolled with a median age of 64 years (range, 36-79) and a median of 4 (range, 3-8) prior lines of therapy. All patients had at least one high risk feature, including failure of prior CAR19 therapy (n=20); refractory disease to second-line or later therapy (n=17); elevated lactate dehydrogenase (LDH) pre-LD (n=17); high tumor burden (n=9); a history of primary refractory disease (n=7); failure of prior autologous hematopoietic stem cell transplantation (HSCT) (n=6); never achieving CR to any therapy (n=5); or LBCL with MYC gene rearrangements (n=5). Successful manufacturing of cells was achieved in all patients. All patients reached day 28 post-infusion and are included in the safety and efficacy analysis presented here; updated results will be presented at the meeting. Every patient experienced cytokine release syndrome (CRS); 20/21 (95%) were Grade 1-2, 1/21 (5%) were Grade 3. Four patients (19%) experienced immune effector cell-associated neurotoxicity syndrome (ICANS); all cases were Grade 1-2 and resolved within 2 days. Five patients (24%) experienced a hyperinflammatory macrophage activation syndrome (MAS), manifested in all cases by pancytopenia and consumptive coagulopathy (DIC) requiring transfusion and/or growth factor support. One patient who received DL2 had a Grade 5 infectious event in the setting of ongoing MAS and pancytopenia. Relative to DL1, higher prevalence of Grade ≥3 cytopenias beyond D28 (89% vs. 50%) and MAS (33% vs. 17%) were observed at DL2; thus, DL1 was selected as the maximally tolerated dose (MTD). ORR at D28 was 86% (CR, n=11; PR, n=7), and was similar between DL1 and DL2 (92% vs. 78%; p=ns). 3/7 (43%) initial PR improved to CR at a median of 3 months post-infusion. All 14 patients (67% of cohort) who achieved CR remain in remission, with a mean follow-up of 7.3 months (range, 1.2-21.3); median progression free survival (PFS) and OS have not yet been reached. Five patients died from disease progression, and one patient died from septic shock in CR. CD22 expression by flow was downregulated or absent in 1/3 (33%) patients evaluated after relapse. Peak CAR-T expansion as detected by peripheral blood flow cytometry occurred at a median of 14 days, with a trend towards earlier and higher peak levels in DL2 patients. Significantly higher mean CAR-T levels occurred at peak expansion in patients who developed MAS (1070±915 vs. 196±209 CAR+ cells/μL; p=0.001). CONCLUSIONS: Infusion of CAR22 in R/R LBCL is safe and well tolerated at DL1. Manufacturing of CAR22 was uniformly successful. With a mean follow-up of 7.3 months, the ORR and CR rates are 18/21 (86%) and 14/21 (67%), respectively. These data demonstrate CAR22 to be an effective salvage therapy for CAR19-refractory or CD19-negative LBCL. Figure 1 Figure 1. Disclosures Frank: Allogene Therapeutics: Research Funding; Adaptive Biotechnologies: Research Funding; Kite-Gilead: Membership on an entity's Board of Directors or advisory committees. Oak: Kite Pharma-Gilead: Research Funding. Arai: Magenta Therapeutics: Research Funding. 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. Shiraz: Kite Pharma-Gilead: Research Funding. Sidana: Janssen: Consultancy, Research Funding; Allogene: Research Funding; Magenta Therapeutics: Consultancy, Research Funding; BMS: Consultancy. Weng: Kite Pharma: Research Funding. Davis: Novartis Pharmaceuticals: Honoraria; Jazz Pharmaceuticals: Research Funding. Feldman: Samsara Biocapital: Consultancy; Obsidian: Consultancy; Lonza PerMed: Consultancy; Gradalis: Consultancy. Mackall: Lyell: Consultancy, Current equity holder in publicly-traded company, Other: Founder; Syncopation Life Sciences: Consultancy, Current holder of individual stocks in a privately-held company, Other: Founder; Apricity: Consultancy, Current equity holder in publicly-traded company; Neoimmune Tech: Consultancy; Nektar: Consultancy, Research Funding. Miklos: Kite, a Gilead Company, Amgen, Atara, Wugen, Celgene, Novartis, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Bioscience, Adicet, Pharmacyclics, Janssen, Takeda, Adaptive Biotechnologies and Miltenyi Biotechnologies: Consultancy; Pharmacyclics: Patents & Royalties; Pharmacyclics, Amgen, Kite, a Gilead Company, Novartis, Roche, Genentech, Becton Dickinson, Isoplexis, Miltenyi, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Biosciences, Adicet, Adaptive Biotechnologies: Research Funding; Adaptive Biotechnologies, Novartis, Juno/Celgene-BMS, Kite, a Gilead Company, Pharmacyclics-AbbVie, Janssen, Pharmacyclics, AlloGene, Precision Bioscience, Miltenyi Biotech, Adicet, Takeda: Membership on an entity's Board of Directors or advisory committees. Muffly: Astellas, Jasper, Adaptive, Baxalta: Research Funding; Adaptive: Honoraria, Other: fees for non-CME/CE services: , Research Funding; Pfizer, Amgen, Jazz, Medexus, Pfizer: Consultancy. OffLabel Disclosure: CD22-directed CAR-T therapy for the treatment of adults with relapsed/refractory large B-cell lymphoma
- Published
- 2021
48. Orca-T Results in High Gvhd-Free and Relapse-Free Survival Following Myeloablative Conditioning for Hematological Malignancies: Results of a Single Center Phase 2 and a Multicenter Phase 1b Study
- Author
-
Caspian Oliai, Parveen Shiraz, Wen-Kai Weng, Andrew R. Rezvani, Joseph P. McGuirk, Robert S. Negrin, Bronwen E. Shaw, Rasmus T. Hoeg, Arpita Gandhi, J Scott McClellan, Sally Arai, Everett Meyer, Vaibhav Agrawal, Rohtesh S. Mehta, Gerhard Bauer, Laura Johnston, David B. Miklos, Amy Putnam, Samer A. Srour, John S. Tamaresis, Mehrdad Abedi, Ying Lu, Anna Moroz, Lori Muffly, Matthew J. Frank, Robert Lowsky, Nathaniel Fernhoff, and Edmund K. Waller
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Myeloablative conditioning ,Immunology ,Cell Biology ,Hematology ,Single Center ,Biochemistry ,Relapse free survival ,Internal medicine ,Phase (matter) ,Medicine ,business - Abstract
BACKGROUND GVHD and non-relapse mortality (NRM) remain frequent complications of HLA-matched HSCT despite the use of standard immunosuppression like tacrolimus and methotrexate. Alternative GVHD prophylaxis (PPX) strategies like T-cell depletion and post-transplant cyclophosphamide negatively impact relapse, infection, and organ toxicity, and no strategy has yet demonstrated a clear benefit for GVHD-free survival. Orca-T is an investigational cellular product comprising stem and immune cells that leverages highly purified donor regulatory T cells to control alloreactive immune responses. Unlike point-of-care graft engineering approaches, Orca-T is produced in a central GMP laboratory and has been successfully distributed to multiple centers across in the U.S. Early clinical trials using Orca-T showed a good safety profile, promising GVHD control, and potentially improved immune reconstitution. Here, we present trial results from both a single-institution Phase 1/2 trial that has completed enrollment and an ongoing multicenter Phase 1b trial. METHODS As of 28 July 2021, 113 patients aged 18-72 have received Orca-T for AML, ALL, MDS, lymphoma, or myelofibrosis. We present here data from 80 patients that have ≥90 days follow-up. 28 and 52 patients, respectively, received Orca-T followed by single-agent GVHD prophylaxis on a single-center Phase 2 study (NCT01660607) and a multicenter Phase Ib (NCT04013685). Orca-T products were derived from HLA-matched related (n=46) or unrelated (n=34) donors. Patients received a variety of myeloablative conditioning regimens (e.g., non-TBI, n=66; TBI-based, n=14) followed by single-agent PPX with either tacrolimus (n=73) or sirolimus (n=7). Median follow-up for these patients is 541 days (single-center) and 248 days (multicenter). We identified a contemporaneous SOC cohort, and we reported on their clinical outcomes at Stanford (n=95) with both matched related (n=52) and unrelated (n=43) transplant recipients who received unmanipulated PBSC products (median f/u 546) and methotrexate plus tacrolimus prophylaxis. RESULTS The Orca-T investigational cell therapy was manufactured reliably, delivered in less than 72 hours for all patients, and every patient enrolled received Orca-T. The Treg drug product was characterized by high Treg purity of 93.8% +/- 3.1% and a dose of 2.6 +/- 0.4 x 106 per kg (equivalent between trials). An Orca-T product was produced and infused for all patients, and there were no logistics failures or infusion reactions. All patients engrafted and Orca-T patients showed earlier neutrophil (median of 12 days vs. 14 days, p On the single-center, Phase 2 clinical trial study at Stanford there is evidence of improved 1-year GVHD and relapse-free survival (GRFS) which was 77% (CI 51-88%) for Orca-T patients vs 34% (CI 25-44%) with SOC (Figure 1A). We observed improved rates of >grade 2 acute GVHD at Day +180 (aGVHD, 14% versus 33%), moderate-to-severe chronic GVHD at 1 year (4% versus 42%) and NRM at 1 year (0% versus 13%). Relapse-free (RFS) and overall survival (OS) trended upwards for Orca-T. Severe infectious complications were rare. Key clinical results from both Orca-T trials are summarized in Table 1; 23 of 80 patients had ≥1 year follow-up. Consistent with findings from the single-institution study, on the multicenter study, rates of moderate-to-severe cGVHD and non-relapse mortality were low at 1-year post-transplant at 3% and 4%, respectively. For all patients who received Orca-T across both studies, we observed GRFS of 72% (Figure 1B), RFS of 78%, and OS of 91% at 1 year. These survival rates compare favorably to the contemporaneous SOC control (33%, 71% and 78%, respectively). Immune reconstitution in Orca-T patients with single agent tacrolimus appears similar to SOC except for observable differences in the IL-2 pathway. CONCLUSIONS Manufacture of high precision Orca-T investigational cell therapy drug products was scaled in a central GMP with reliable distribution to centers. Patients that received Orca-T and single-agent PPX showed significantly reduced aGVHD, cGVHD and NRM. Orca-T shows promise to improve GRFS and other transplant outcomes. Orca-T has been granted Regenerative Medicine Advanced Therapy status by the FDA, and a phase 3 prospectively, randomized study is planned. Figure 1 Figure 1. Disclosures Gandhi: Gamida Cell: Consultancy, Membership on an entity's Board of Directors or advisory committees; CareDx Inc: Honoraria. Muffly: Pfizer, Amgen, Jazz, Medexus, Pfizer: Consultancy; Astellas, Jasper, Adaptive, Baxalta: Research Funding; Adaptive: Honoraria, Other: fees for non-CME/CE services: , Research Funding. Shiraz: Kite Pharma-Gilead: Research Funding. Mehta: Kadmon: Research Funding; Incyte: Research Funding; CSLBehring: Research Funding; Syndax: Research Funding. McGuirk: Allovir: Consultancy, Honoraria, Research Funding; Novartis: Research Funding; Magenta Therapeutics: Consultancy, Honoraria, Research Funding; Gamida Cell: Research Funding; Kite/ Gilead: Consultancy, Honoraria, Other: travel accommodations, expense, Kite a Gilead company, Research Funding, Speakers Bureau; Juno Therapeutics: Consultancy, Honoraria, Research Funding; EcoR1 Capital: Consultancy; Bellicum Pharmaceuticals: Research Funding; Pluristem Therapeutics: Research Funding; Fresenius Biotech: Research Funding; Astelllas Pharma: Research Funding; Novartis: Research Funding. Waller: Verastem Oncology: Consultancy, Research Funding; Cambium Oncology: Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Arai: Magenta Therapeutics: Research Funding. Rezvani: US Department of Justice: Consultancy; Nohla Therapeutics: Other: One-time scientific advisory board; Pharmacyclics-Abbvie: Research Funding; Kaleido: Other: One-time scientific advisory board. Weng: Kite Pharma: Research Funding. Miklos: Adaptive Biotechnologies, Novartis, Juno/Celgene-BMS, Kite, a Gilead Company, Pharmacyclics-AbbVie, Janssen, Pharmacyclics, AlloGene, Precision Bioscience, Miltenyi Biotech, Adicet, Takeda: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics, Amgen, Kite, a Gilead Company, Novartis, Roche, Genentech, Becton Dickinson, Isoplexis, Miltenyi, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Biosciences, Adicet, Adaptive Biotechnologies: Research Funding; Kite, a Gilead Company, Amgen, Atara, Wugen, Celgene, Novartis, Juno-Celgene-Bristol Myers Squibb, Allogene, Precision Bioscience, Adicet, Pharmacyclics, Janssen, Takeda, Adaptive Biotechnologies and Miltenyi Biotechnologies: Consultancy; Pharmacyclics: Patents & Royalties. Frank: Kite-Gilead: Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Research Funding; Allogene Therapeutics: Research Funding. Fernhoff: Orca Bio: Current Employment. Putnam: Orca Bio: Current Employment. McClellan: Orca Bio: Current Employment, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Shaw: Orca bio: Consultancy; mallinkrodt: Other: payments. Abedi: Seattle Genetics: Speakers Bureau; BMS/Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Speakers Bureau. Meyer: Triursus Therapeutics: Current holder of stock options in a privately-held company; GigaImmune: Current holder of stock options in a privately-held company; Orca Biosystems: Research Funding; Indee, Jura: Consultancy.
- Published
- 2021
49. Validation of the Hematopoietic Cell Transplantation–Specific Comorbidity Index in Nonmyeloablative Allogeneic Stem Cell Transplantation
- Author
-
Lingyao Yang, David B. Miklos, Robert S. Negrin, Everett Meyer, Laura Johnston, Andrew R. Rezvani, Robert Lowsky, Lori Muffly, Judith A. Shizuru, Vandana Sundaram, Sally Arai, Wen-Kai Weng, and Muthu Veeraputhiran
- Subjects
Adult ,medicine.medical_specialty ,Transplantation Conditioning ,Allogeneic transplantation ,Comorbidity ,Risk Assessment ,Article ,Pulmonary function testing ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Diabetes mellitus ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Aged ,Retrospective Studies ,Preparative Regimen ,Transplantation ,business.industry ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Confidence interval ,Surgery ,Survival Rate ,Regimen ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Regression Analysis ,business ,030215 immunology - Abstract
The Hematopoietic Cell Transplantation (HCT)–Specific Comorbidity Index (HCT-CI) has been extensively studied in myeloablative and reduced-intensity conditioning regimens, with less data available regarding the validity of HCT-CI in nonmyeloablative (NMA) allogeneic transplantation. We conducted a retrospective analysis to evaluate the association between HCT-CI and nonrelapse mortality (NRM) and all-cause mortality (ACM) in patients receiving the total lymphoid irradiation and antithymocyte globulin (TLI/ATG) NMA transplantation preparative regimen. We abstracted demographic and clinical data from consecutive patients, who received allogeneic HCT with the TLI/ATG regimen between January 2008 and September 2014, from the Stanford blood and marrow transplantation database. We conducted univariable and multivariable Cox proportional hazards regression models to evaluate the association between HCT-CI and NRM and ACM. In all, 287 patients were included for analysis. The median age of the patients was 61 (range, 22 to 77) years. The median overall survival was 844 (range, 374 to 1484) days. Most patients had Karnofsky performance score of 90 or above (85%). Fifty-two (18%) patients relapsed within 3 months and 108 (38%) patients relapsed within 1 year, with a median time to relapse of 163 (range, 83 to 366) days. Among the comorbidities in the HCT-CI identified at the time of HCT, reduced pulmonary function was the most common (n = 89), followed by prior history of malignancy (n = 39), psychiatric condition (n = 38), and diabetes (n = 31). Patients with higher HCT-CI scores had higher mortality risks for ACM (hazard ratio [HR], 1.95; 95% confidence interval [CI], 1.22 to 3.14 for HCT-CI score 1 or 2 and HR, 1.85; 95% CI, 1.11 to 3.08 for HCT-CI score ≥ 3, compared with 0, respectively). Among individual HCT-CI variables, diabetes (HR, 2.31; 95% CI, 1.79 to 2.89; P = .003) and prior solid tumors (HR, 1.75; 95% CI, 1.02 to 3.00; P = .043) were associated with a higher risk of ACM. Higher HCT-CI scores were significantly associated with higher risk of death. HCT-CI is a valid tool for predicting ACM in NMA TLI/ATG allogeneic HCT.
- Published
- 2017
50. Survival Following Post-HCT Relapse in Adult Acute Lymphoblastic Leukemia Has Improved in the Era of Novel Immunotherapies: A Single Institution Analysis
- Author
-
Matthew J. Frank, Maria Iglesias, Robert S. Negrin, Juliana Craig, Andrew R. Rezvani, Judith A. Shizuru, Everett Meyer, Parveen Shiraz, Robert Lowsky, Surbhi Sidana, Laura Johnston, David B. Miklos, Kristen M. Cunanan, Lori Muffly, Sally Arai, and W. K. Weng
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Immunology ,Adult Acute Lymphoblastic Leukemia ,medicine ,Cell Biology ,Hematology ,Single institution ,business ,Biochemistry - Abstract
Background: Historically, the survival of adult patients with acute lymphoblastic leukemia (ALL) relapsing after allogeneic hematopoietic cell transplantation (HCT) was dismal, with fewer than 10% surviving long-term (Fielding et al Blood 2007). In the last five years, the availability of targeted immunotherapies including blinatumomab (blin), inotuzumab ozogamicin (IO), and chimeric antigen T-cell receptor (CART) therapy has expanded the opportunity for effective salvage of relapsed/refractory ALL. We hypothesized that the expanding therapeutic landscape has resulted in superior survival following post-HCT relapse in adult ALL in the targeted immunotherapy era. Methods: We performed a retrospective analysis of adults receiving first allogeneic HCT for ALL between 2008-2019 at Stanford University; patients were stratified by time period of HCT: 2008-2013 (earlier era) vs 2014-2019 (recent immunotherapy era). Descriptive statistics characterized the study cohort; chi-square test was used to evaluate differences in characteristics and treatments based on time period transplanted. Kaplan Meier method was used to determine overall survival (OS); log-rank tested significance between time periods. Follow-up time of the earlier time period was truncated to 5.4 years to match maximum follow-up time of the more recent time period. Results: Of the 285 adult ALL patients transplanted (N=119, 2008-2013; N=166, 2014-2019), 81 (28%) experienced disease relapse following HCT and represent the analytic cohort. Post-HCT relapse occurred in 39 (33%) transplanted between 2008-2013 and 42 (25%) transplanted between 2014-2019. The median time to relapse following HCT was 7.72 months (95% CI, 3.88-14.0), and did not significantly differ between time periods. Baseline patient and transplant characteristics were similar across the two time periods (Table 1); however, relative to the earlier time period, patients transplanted during the more recent time period were less likely to be transplanted with active disease (26% vs 2%), and more likely to receive cord blood as a stem cell source (0 vs 10%). The median overall survival (OS) for the entire cohort following post-HCT relapse was 9.93 months (95% CI, 7.07-12.73). However, the median OS from relapse was 7.99 months (95% CI, 3.30-11.6) for patients transplanted in the earlier era, while the median OS for patients relapsing in the recent era was 15.75 months (95% CI, 9.24-26.4), P Conclusion: In this large cohort of adults transplanted for ALL over the last decade, we show that the OS of ALL patients relapsing after HCT has significantly improved, coinciding with a substantial increase in the availability and utilization of novel therapies in this setting. Additional studies are needed to understand the optimal therapeutic intervention for post-HCT relapse in adult ALL. Disclosures Meyer: Orca Bio: Research Funding. Miklos:Adaptive Biotech: Consultancy, Other: Travel support, Research Funding; Kite-Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel support, Research Funding; Juno-Celgene-Bristol-Myers Squibb: Consultancy, Other: Travel support, Research Funding; Allogene Therapeutics Inc.: Research Funding; Novartis: Consultancy, Other: Travel support, Research Funding; Pharmacyclics: Consultancy, Other: Travel support, Patents & Royalties, Research Funding; Janssen: Consultancy, Other: Travel support; Miltenyi Biotec: Research Funding. Negrin:UpToDate: Honoraria; Biosource: Current equity holder in private company; Amgen: Consultancy; BioEclipse Therapeutics: Current equity holder in private company; Magenta Therapeutics: Consultancy, Current equity holder in publicly-traded company; KUUR Therapeutics: Consultancy. Rezvani:Pharmacyclics: Research Funding. Shizuru:Jasper Therapeutics, Inc: Current equity holder in private company, Membership on an entity's Board of Directors or advisory committees. Sidana:Janssen: Consultancy. Shiraz:ORCA BioSystems: Research Funding; Kite, a Gilead Company: Research Funding. Muffly:Amgen: Consultancy; Adaptive: Research Funding; Servier: Research Funding.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.