205 results on '"George Carrum"'
Search Results
2. Allogeneic, off-the-shelf, SARS-CoV-2-specific T cells (ALVR109) for the treatment of COVID-19 in high-risk patients
- Author
-
Spyridoula Vasileiou, LaQuisa Hill, Manik Kuvalekar, Aster G. Workineh, Ayumi Watanabe, Yovana Velazquez, Suhasini Lulla, Kimberly Mooney, Natalia Lapteva, Bambi J. Grilley, Helen E. Heslop, Cliona M. Rooney, Malcolm K. Brenner, Todd N. Eagar, George Carrum, Kevin A. Grimes, Ann M. Leen, and Premal Lulla
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Defects in T-cell immunity to SARS-CoV-2 have been linked to an increased risk of severe COVID-19 (even after vaccination), persistent viral shedding and the emergence of more virulent viral variants. To address this T-cell deficit, we sought to prepare and cryopreserve banks of virus-specific T cells, which would be available as a partially HLA-matched, off-the-shelf product for immediate therapeutic use. By interrogating the peripheral blood of healthy convalescent donors, we identified immunodominant and protective T-cell target antigens, and generated and characterized polyclonal virus-specific T-cell lines with activity against multiple clinically important SARS-CoV-2 variants (including ‘delta’ and ‘omicron’). The feasibility of making and safely utilizing such virus-specific T cells clinically was assessed by administering partially HLA-matched, third-party, cryopreserved SARS-CoV-2-specific T cells (ALVR109) in combination with other antiviral agents to four individuals who were hospitalized with COVID-19. This study establishes the feasibility of preparing and delivering off-the-shelf, SARS-CoV-2-directed, virus-specific T cells to patients with COVID-19 and supports the clinical use of these products outside of the profoundly immune compromised setting (ClinicalTrials.gov number, NCT04401410).
- Published
- 2022
- Full Text
- View/download PDF
3. Fourth Annual Summer Research Summit on Health Equity Organized by the Center of Excellence in Health Equity, Training and Research, Baylor College of Medicine, Houston, Texas 77030, USA on May 20, 2021
- Author
-
Aanand Naik, Abbhirami Rajagopal, Adam Floyd, Adriana Gil, Aisha Tepede, Aisha Koroma, Aisha Deslandes, Akua Graf, Alejandra Ruiz-Velasco, Alexa Reyna-Carrillo, Alexandra Alvarenga, Alexia Awoseyi, Alexis Hernandez, Alexis Lawrence, Ali Asghar-Ali, Allyssa Abacan, Alyce Adams, Alyna Khan, Alyson McGregor, Alyssa Hansen, Amari Johnson, Andrea Coj, Andrea Vick, Andria Tatem, Anjali Aggarwal, Anjali Deendyal, Ann Blake, Annabella Awazi, Anne VanHorn, Anuj Marathe, Anusha Jayaram, April Adams, Arabella Hall, Ariana Heredia, Ariana Chavarria, Asha Morrow, Ashley Butler, Asia Hodges, Aura Mejia, Avani Patel, Ayleen Hernandez, Benjamin Akande, Blessing Felix-Okoroji, Brisa Garcia, Buckleitner Jenna, Callie Fischer, Camden Hallmark, Cara Coren, Carlos Ramos, Cecilia Gambala, Charleta Guillory, Chelsea Livingston, Chioma Onyejiaka, Chishinga Callender, Christina Aldrich, Christopher Largaespada, Claire Bocchini, Craig Cochran, Danielle Sherman, Danielle Gonzales, David Venzon, David Wittkower, Debbe Thompson, Deborah Thompson, Debra Eseonu, Deepa Dongarwar, Delia Rospigliosi, Denise Smart, Denisse Velazquez, Derek Lockett, Eberechi Nwogu-Onyemkpa, Elizabeth Byrne, Elyse Lopez, Eric Dybbro, Eric Storch, Erica Onwuegbuchu, Erica Valdes, Erin Donovan, Eunique Williams, Evan Keil, Faith Ihekweazu, Felicia Rosiji, Gabriela Espinoza-Candelaria, Gabriella Chmaitelli, Gabriella Tavera, Gail Oneal, Gal Barbut, Gauvain Tonpouwo, George Carrum, Gina DeFelice, Hamisu Salihu, Heather Haq, Helen Heslop, Houston Lester, Ifeoma Ezenwabachili, Ila Gautham, Jacquelin Powell, Jaime Alleyn, Jasmine King, Jaydira Rivero, Jayer Chung, Jayna Dave, Jean Raphael, Jen Bryan, Jendi Haug, Jennifer Bryan, Jenny Blau, Jerry Bellamy, Jessica Medrano, Jessica Ramirez, Jocelyn Greely, Jonnae Atkinson, Jorge Miranda, Jose Dominguez, Jose Roca, Joseph Mills, Joshua Hamer, Joshua Muñiz, Julliet Ogu, Karen Gibbs, Karen Johnson, Karen Riggins, Karla Fredricks, Keila Lopez, Kellie Williams, Keyishi Peters, Kil Hyein, LaQuisa Hill, Lee Weinstein, Lena Shay, Lentz Lefevre, Lindy Ross, Lisa Noll, Lois Akpati, Lorin Crear, Lucy Puryear, Maame Coleman, Madhuri Vasudevan, Malachi Miller, Maria Vigil-Mallette, Maria Jaramillo, Mariam Chacko, Mariana Baroni, Mariana Murillo, Maricarmen Marroquin, Marina Masciale, Marlene McNeese, Martinez Austin, Matthew Koller, Maya Lee, Maziar Nourian, Megan Abadom, Meghna Sebastian, Meheret Adera, Mei-Lei Laracuente, Michelle Lopez, Michelle Wright, Miguel Montero-Baker, Monica Gonzalez, Morrow Adelene, Mosope Adeyeye, Muzaffar Qazilbash, Namrata Walia, Nancy Shenoi, Natalia Rodriguez, Naya Mukdadi, Neeraj Saini, Norma Olvera, Ololade Chris-Rotimi, Paige Hoyer, Parisa Fallah, Peggy Smith, Premal Lulla, Priscilla Ehieze, Priyanka Murali, Rachel Head, Rachel Nwaneri, Rachelle Wanser, Racquel Lyn, Rammurti Kamble, Ramyar Gilani, Raquel Martinez, Rathi Asaithambi, Reginald Hatter, Rhanna Wilson, Ria Brown, Robert Shulman, Robert Mbilinyi, Robert Levine, Roe Avery, Romil Patel, Roslyn Aduhene-Opoku, Ruth Mizu, Saad Usmani, Sadia Usmani, Saeed Ahmed, Samantha Moore, Samer Hadidi, Sana Erabti, Sana Javed, Sana Younus, Sanders Mar’Quenda, Sandy Samaan, Sara Alam, Sara Welty, Sergio Navarro, Shad Deering, Shaine Morris, Shana Alford, Shangir Siddique, Shantyka Walton, Shayan Bhathena, Shelease O’Bryant, Shital Patel, Sindhu Idicula, Sophia Banu, Sophie Albert, Sophie McCullum, Sophie Lin, Star Okolie, Sunita Agarwal, Susan Gillespie, Syed Hussaini, Sylvia Hysong, Tammy Kang, Tara Everett, Taylor Ottesen, Tiana DiMasi, Tien Nguyen, Toi Harris, Tzu-An Chen, Vicki Mercado, Victoria Michael, Victoria Xie, William Simonds, Yesenya Gonzalez, Yicenia Aviles, Ynhi Thomas, and Zachary Pallister
- Subjects
Public aspects of medicine ,RA1-1270 - Abstract
The fourth annual summer research summit organized by the Center of Excellence (COE) in Health Equity, Training and Research, Baylor College of Medicine (BCM) was held on May 20, 2021. The theme of this year’s summit was ‘Strengthening Our Commitment to Racial and Social Justice to Improve Public Health.’ Given the ongoing pandemic, the summit was conducted virtually through digital platforms. This program was intended for both BCM and external audiences interested in advancing health equity, diversity and inclusion in healthcare among healthcare providers and trainees, biomedical scientists, social workers, nurses, individuals involved in talent acquisition and development such as hiring managers (HR professionals), supervisors, college and hospital affiliate leadership and administrators, as well as diversity and inclusion excellence practitioners. We had attendees from all regions of the United States, India, Pakistan and the Demographic Republic of the Congo. The content in this Book of Abstracts encapsulates a summary of the research efforts by the BCM COE scholars (which includes post-baccalaureate students, medical students, clinical fellows and junior faculty from BCM) as well as the external summit participants. The range of topics in this year’s summit was quite diverse encompassing disparities in relation to maternal and child health (MCH), immigrant heath, cancers, vaccination uptakes and COVID-19 infections. Various solutions were ardently presented to address these disparities including community engagement and partnerships, improvement in health literacy and development of novel technologies and therapeutics. With this summit, BCM continues to build on its long history of educational outreach initiatives to promote diversity in medicine by focusing on programs aimed at increasing the number of diverse and highly qualified medical professionals ready to introduce effective and innovative approaches to reduce or eliminate health disparities. These programs will improve information resources, clinical education, curricula, research and cultural competence as they relate to minority health issues and social determinants of health. The summit received very positive response in terms of zealous participation and outstanding evaluations; and overall, it was a great success. Copyright © 2021 Lopez et al. Published by Global Health and Education Projects, Inc. This is an open-access article distributed under the terms of the Creative Commons Attribution License CC BY 4.0.
- Published
- 2021
- Full Text
- View/download PDF
4. Supplementary Tables 1 - 2 from Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity
- Author
-
Catherine M. Bollard, Malcolm K. Brenner, Helen E. Heslop, Kathryn Leung, Robert A. Krance, Carlos A. Ramos, George Carrum, Eric Yvon, Barbara Savoldo, Aaron Foster, Sawa Ito, A. John Barrett, Jos Melenhorst, Hao Liu, Meng-Fen Wu, Yasmin Hazrat, Paul Castillo-Caro, Stephanie Ku, and Alana A. Kennedy-Nasser
- Abstract
PDF file - 63KB, Supplemental Table 1. Recipients of MRD Grafts who did not receive ULD-IL-2 Supplemental Table 2. Recipients of MUD Grafts who did not receive ULD-IL-2.
- Published
- 2023
5. Data from Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity
- Author
-
Catherine M. Bollard, Malcolm K. Brenner, Helen E. Heslop, Kathryn Leung, Robert A. Krance, Carlos A. Ramos, George Carrum, Eric Yvon, Barbara Savoldo, Aaron Foster, Sawa Ito, A. John Barrett, Jos Melenhorst, Hao Liu, Meng-Fen Wu, Yasmin Hazrat, Paul Castillo-Caro, Stephanie Ku, and Alana A. Kennedy-Nasser
- Abstract
Purpose: GVHD after allogeneic hematopoietic stem cell transplantation (alloSCT) has been associated with low numbers of circulating CD4+CD25+FoxP3+ regulatory T cells (Tregs). Because Tregs express high levels of the interleukin (IL)-2 receptor, they may selectively expand in vivo in response to doses of IL-2 insufficient to stimulate T effector T-cell populations, thereby preventing GVHD.Experimental Design: We prospectively evaluated the effects of ultra low-dose (ULD) IL-2 injections on Treg recovery in pediatric patients after alloSCT and compared this recovery with Treg reconstitution post alloSCT in patients without IL-2. Sixteen recipients of related (n = 12) or unrelated (n = 4) donor grafts received ULD IL-2 post hematopoietic stem cell transplantation (HSCT; 100,000–200,000 IU/m2 ×3 per week), starting Results: No grade 3/4 toxicities were associated with ULD IL-2. CD4+CD25+FoxP3+ Tregs increased from a mean of 4.8% (range, 0%–11.0%) pre IL-2 to 11.1% (range, 1.2%–31.1%) following therapy, with the greatest change occurring in the recipients of matched related donor (MRD) transplants. No IL-2 patients developed grade 2–4 acute GVHD (aGVHD), compared with 4 of 33 (12%) of the comparator group who did not receive IL-2. IL-2 recipients retained T cells reactive to viral and leukemia antigens, and in the MRD recipients, only 2 of 13 (15%) of the IL-2 patients developed viral infections versus 63% of the comparator group (P = 0.022).Conclusions: Hence, ULD IL-2 is well tolerated, expands a Treg population in vivo, and may be associated with a lower incidence of viral infections and GVHD. Clin Cancer Res; 20(8); 2215–25. ©2014 AACR.
- Published
- 2023
6. Supplementary Figure 1 from Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity
- Author
-
Catherine M. Bollard, Malcolm K. Brenner, Helen E. Heslop, Kathryn Leung, Robert A. Krance, Carlos A. Ramos, George Carrum, Eric Yvon, Barbara Savoldo, Aaron Foster, Sawa Ito, A. John Barrett, Jos Melenhorst, Hao Liu, Meng-Fen Wu, Yasmin Hazrat, Paul Castillo-Caro, Stephanie Ku, and Alana A. Kennedy-Nasser
- Abstract
PDF file - 26KB, General Immune reconstitution. Immune reconstitution was evaluated in individual patients after transplant. Phenotyping was performed on PBMC to evaluate for the presence of NK cells (A), CD8+ T-cells (B), CD4+ T cells (C), B cells (D) and CD56+/CD3+ NK-like T-cells (E).
- Published
- 2023
7. Rituximab as adjunctive therapy to BEAM conditioning for autologous stem cell transplantation in Hodgkin lymphoma
- Author
-
Brian D. Friend, Ibrahim N. Muhsen, Shreeya Patel, LaQuisa C. Hill, Premal Lulla, Carlos A. Ramos, S. Ravi Pingali, Rammurti T. Kamble, Tami D. John, Baheyeldin Salem, Saleh Bhar, Erin E. Doherty, John Craddock, Ghadir Sasa, Mengfen Wu, Tao Wang, Caridad Martinez, Robert A. Krance, Helen E. Heslop, and George Carrum
- Subjects
Transplantation ,Hematology - Published
- 2022
8. Efficacy of Afternoon Plerixafor Administration for Stem Cell Mobilization
- Author
-
Cynthia El Rahi, James Eldin Cox, Romelia May, George Carrum, Gloria Obi Anyadike, Audrey Scholoff, and Rammurti Kamble
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background: When used for hematopoietic stem cell mobilization, plerixafor was originally recommended to be administered 11 hours prior to apheresis based on the peak effect of 10 to 14 hours translating into an administration time of 10 to 11 pm . Reports of post-plerixafor anaphylactic reactions mandated labeling change by the Food and Drug Administration with recommendation of monitoring patients after administration. Based on data suggesting sustained plerixafor activity at 18 hours, we changed our administration time to 4 pm at our center. Objective: The objective of this study is to compare the stem cell collection efficiency before and after the practice change at our institution. Methods: A retrospective chart review for patients with multiple myeloma, Hodgkin lymphoma, and non-Hodgkin lymphoma who received a plerixafor-containing mobilization regimen was conducted. The primary end point was the percentage of patients achieving the minimal CD34 + cell goal in ⩽2 apheresis days. The secondary end points included the percentage of patients achieving the preferred CD34 + cell goal in ⩽2 apheresis days, days of apheresis, total CD34 + cells Collected, and engraftment time. Results: A total of 208 patients (4 pm group n = 68, 10 pm group n = 140) with multiple myeloma (n = 112), Hodgkin lymphoma (n = 10), and non-Hodgkin lymphoma (n = 86) were included in the analysis. About 91% and 89% ( P = .804) of the patients in the 4 and 10 pm groups, respectively, collected minimum cell dose. Preferred CD34 + cell goal was achieved in 57% and 53% of patients in the 4 and 10 pm groups, respectively. Conclusions: Late afternoon administration of plerixafor provides efficient stem cell mobilization.
- Published
- 2018
- Full Text
- View/download PDF
9. Health disparities experienced by Black and Hispanic Americans with multiple myeloma in the United States: a population-based study
- Author
-
Deepa Dongarwar, Premal Lulla, Helen E. Heslop, Saad Z. Usmani, Samer Al Hadidi, George Carrum, Rammurti T. Kamble, Carlos A. Ramos, Hamisu M. Salihu, and La Quisa C. Hill
- Subjects
Cancer Research ,business.industry ,Incidence (epidemiology) ,Hispanic or Latino ,Hematology ,medicine.disease ,United States ,White People ,Article ,Health equity ,Black or African American ,Population based study ,Cross-Sectional Studies ,Oncology ,medicine ,Humans ,Multiple Myeloma ,business ,Multiple myeloma ,Demography - Abstract
Hispanics and non-Hispanic (NH)-Blacks continue to face numerous health disparities related to multiple myeloma (MM). We aimed to analyze trends of MM-related hospitalizations and incidence of in-hospital mortality with a 10-year cross-sectional analysis of inpatient hospitalizations. The prevalence of MM-related hospitalizations was higher in NH-Blacks compared to NH-Whites (476.0 vs. 305.6 per 100,000 hospitalizations, p < .001). MM-related in-hospital mortality was higher in Hispanics compared to NH-Whites and NH-Blacks (6.2 vs. 5.3%, p < .001). Using average annual percent change (AAPC), we found a statistically significant decline of in-hospital mortality among all MM patients except NH-Blacks (AAPC: −2.2, 95% confidence interval (CI) −4.7, 0.4, p = .47), who had the highest inpatient mortality in recent years. Multivariate analysis showed that NH-Blacks received fewer transplants, more blood product transfusions, fewer palliative care consults, less inpatient chemotherapy, and utilized more intensive care. Disparities in MM care for NH-Blacks and Hispanics continue to persist despite recent advancements in MM therapy.
- Published
- 2021
10. Protracted Diarrhea from High-Dose Melphalan in Autologous Hematopoietic Transplantation: Response to Short Course of Oral Budesonide
- Author
-
Yugi Zhang, Zimu Gong, James Cox, Breanna Hinman, Shalewa Oki, Susan Jacob, George Carrum, and Rammurti T. Kamble
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
11. Clinical effects of administering leukemia-specific donor T cells to patients with AML/MDS after allogeneic transplant
- Author
-
Tao Wang, Helen E. Heslop, Stephen Gottschalk, Bambi Grilley, Rammurti T. Kamble, Carlos A. Ramos, Ann M. Leen, Meng-Fen Wu, Malcolm K. Brenner, Robert A. Krance, Catherine Robertson, Manik Kuvalekar, Jasleen K. Randhawa, Spyridoula Vasileiou, Adrian P. Gee, Suhasini Lulla, Juan F. Vera, Betty Chung, Ayumi Watanabe, Premal Lulla, Ifigeneia Tzannou, Swati Naik, LaQuisa Hill, and George Carrum
- Subjects
Adult ,Male ,Adolescent ,Clinical Trials and Observations ,T-Lymphocytes ,medicine.medical_treatment ,Immunology ,Graft vs Host Disease ,T-Cell Antigen Receptor Specificity ,Graft vs Leukemia Effect ,Hematopoietic stem cell transplantation ,Biochemistry ,Young Adult ,Antigen ,Antigens, Neoplasm ,Recurrence ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Survivin ,medicine ,Humans ,Transplantation, Homologous ,Aged ,Salvage Therapy ,PRAME ,Errata ,business.industry ,Hematopoietic Stem Cell Transplantation ,Myeloid leukemia ,Cell Biology ,Hematology ,Middle Aged ,Allografts ,Donor Lymphocytes ,medicine.disease ,Combined Modality Therapy ,Tissue Donors ,Leukemia, Myeloid, Acute ,Leukemia ,surgical procedures, operative ,Lymphocyte Transfusion ,Myelodysplastic Syndromes ,Cancer research ,Female ,Stem cell ,business - Abstract
Relapse after allogeneic hematopoietic stem cell transplantation (HCT) is the leading cause of death in patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Infusion of unselected donor lymphocytes (DLIs) enhances the graft-versus-leukemia (GVL) effect. However, because the infused lymphocytes are not selected for leukemia specificity, the GVL effect is often accompanied by life-threatening graft-versus-host disease (GVHD), related to the concurrent transfer of alloreactive lymphocytes. Thus, to minimize GVHD and maximize GVL, we selectively activated and expanded stem cell donor–derived T cells reactive to multiple antigens expressed by AML/MDS cells (PRAME, WT1, Survivin, and NY-ESO-1). Products that demonstrated leukemia antigen specificity were generated from 29 HCT donors. In contrast to DLIs, leukemia-specific T cells (mLSTs) selectively recognized and killed leukemia antigen–pulsed cells, with no activity against recipient's normal cells in vitro. We administered escalating doses of mLSTs (0.5 to 10 × 107 cells per square meter) to 25 trial enrollees, 17 with high risk of relapse and 8 with relapsed disease. Infusions were well tolerated with no grade >2 acute or extensive chronic GVHD seen. We observed antileukemia effects in vivo that translated into not-yet-reached median leukemia-free and overall survival at 1.9 years of follow-up and objective responses in the active disease cohort (1 complete response and 1 partial response). In summary, mLSTs are safe and promising for the prevention and treatment of AML/MDS after HCT. This trial is registered at www.clinicaltrials.com as #NCT02494167.
- Published
- 2021
12. CT-514 Acute Cardiac Tamponade Following Adult Allogeneic Hematopoietic Transplantation
- Author
-
Yuqi Zhang, Zimu Gong, Gloria Obi, Audrey Scholoff, Jacob Cox, Shalewa Oki, George Carrum, and Rammurti Kamble
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
13. Donor-derived multiple leukemia antigen-specific T-cell therapy to prevent relapse after transplant in patients with ALL
- Author
-
Swati Naik, Spyridoula Vasileiou, Ifigeneia Tzannou, Manik Kuvalekar, Ayumi Watanabe, Catherine Robertson, Natalia Lapteva, Wang Tao, Mengfen Wu, Bambi Grilley, George Carrum, Rammurti T. Kamble, LaQuisa Hill, Robert A. Krance, Caridad Martinez, Priti Tewari, Bilal Omer, Stephen Gottschalk, Helen E. Heslop, Malcom K. Brenner, Cliona M. Rooney, Juan F. Vera, Ann M. Leen, and Premal D. Lulla
- Subjects
Adult ,Transplantation ,Leukemia ,Immunology ,Cell- and Tissue-Based Therapy ,Hematopoietic Stem Cell Transplantation ,Transplants ,Graft vs Host Disease ,Cell Biology ,Hematology ,Biochemistry ,Tissue Donors ,Recurrence ,Chronic Disease ,Humans ,Transplantation, Homologous ,Child - Abstract
Hematopoietic stem cell transplant (HSCT) is a curative option for patients with high-risk acute lymphoblastic leukemia (ALL), but relapse remains a major cause of treatment failure. To prevent disease relapse, we prepared and infused donor-derived multiple leukemia antigen–specific T cells (mLSTs) targeting PRAME, WT1, and survivin, which are leukemia-associated antigens frequently expressed in B- and T-ALL. Our goal was to maximize the graft-versus-leukemia effect while minimizing the risk of graft-versus-host disease (GVHD). We administered mLSTs (dose range, 0.5 × 107 to 2 × 107 cells per square meter) to 11 patients with ALL (8 pediatric, 3 adult), and observed no dose-limiting toxicity, acute GVHD or cytokine release syndrome. Six of 8 evaluable patients remained in long-term complete remission (median: 46.5 months; range, 9-51). In these individuals we detected an increased frequency of tumor-reactive T cells shortly after infusion, with activity against both targeted and nontargeted, known tumor-associated antigens, indicative of in vivo antigen spreading. By contrast, this in vivo amplification was absent in the 2 patients who experienced relapse. In summary, infusion of donor-derived mLSTs after allogeneic HSCT is feasible and safe and may contribute to disease control, as evidenced by in vivo tumor-directed T-cell expansion. Thus, this approach represents a promising strategy for preventing relapse in patients with ALL.
- Published
- 2021
14. Rituximab as adjunctive therapy to BEAM conditioning for autologous stem cell transplantation in Hodgkin lymphoma
- Author
-
Brian D, Friend, Ibrahim N, Muhsen, Shreeya, Patel, LaQuisa C, Hill, Premal, Lulla, Carlos A, Ramos, S Ravi, Pingali, Rammurti T, Kamble, Tami D, John, Baheyeldin, Salem, Saleh, Bhar, Erin E, Doherty, John, Craddock, Ghadir, Sasa, Mengfen, Wu, Tao, Wang, Caridad, Martinez, Robert A, Krance, Helen E, Heslop, and George, Carrum
- Subjects
Adult ,Transplantation Conditioning ,Adolescent ,Cytarabine ,Hematopoietic Stem Cell Transplantation ,Middle Aged ,Carmustine ,Hodgkin Disease ,Transplantation, Autologous ,Young Adult ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Neoplasm Recurrence, Local ,Child ,Rituximab ,Melphalan ,Aged ,Retrospective Studies - Abstract
While high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) leads to improved disease-free survival (DFS) for children and adults with relapsed/refractory Hodgkin lymphoma (HL), relapse remains the most frequent cause of mortality post-transplant. Rituximab has been successfully incorporated into regimens for other B-cell lymphomas, yet there have been limited studies of rituximab in HL patients. We hypothesized that adding rituximab to BEAM (carmustine, etoposide, cytarabine, melphalan) conditioning would reduce relapse risk in HL patients post-transplant. Here, we retrospectively review the outcomes of patients with relapsed/refractory HL who received rituximab in addition to BEAM. The primary outcome was DFS. Our cohort included 96 patients with a median age of 28 years (range, 6-76). Majority of patients (57%) were diagnosed with advanced (Stage III-IV) disease, and 62% were PET negative pre-transplant. DFS was 91.5% at 1 year [95% CI 86-98%], and 78% at 3 years [95% CI 68-88%]. NRM was 0% and 3.5% at 1-year [95% CI 0-3%] and 3-years [95% CI 0-8.5%], respectively. 25% of patients developed delayed neutropenia, with 7% requiring infection-related hospitalizations, and one death. We have demonstrated excellent outcomes for patients receiving rituximab with BEAM conditioning for relapsed/refractory HL. Future comparative studies are needed to better determine whether rituximab augments outcomes post-transplant.
- Published
- 2021
15. Poster: CT-514 Acute Cardiac Tamponade Following Adult Allogeneic Hematopoietic Transplantation
- Author
-
Yuqi Zhang, Zimu Gong, Gloria Obi, Audrey Scholoff, Jacob Cox, Shalewa Oki, George Carrum, and Rammurti Kamble
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
16. CT-207 Protracted Diarrhea from High-Dose Melphalan in Autologous Hematopoietic Transplantation: Response to Short Course of Oral Budesonide
- Author
-
Zimu Gong, Yuqi Zhang, Gloria Obi, Audrey Scholoff, James Cox, Susan Jacob, Shalewa Oki, George Carrum, and Rammurti Kamble
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
17. In Vivo Fate and Activity of Second- versus Third-Generation CD19-Specific CAR-T Cells in B Cell Non-Hodgkin’s Lymphomas
- Author
-
Catherine Robertson, Amy Reyna, Hao Liu, Birju Mehta, Huimin Zhang, Barbara Savoldo, Neeharika Narala, Olga Dakhova, George Carrum, Adrian P. Gee, Zhuyong Mei, Meng Fen Wu, Gianpietro Dotti, Helen E. Heslop, Rammurti T. Kamble, Bambi Grilley, Malcolm K. Brenner, Rayne H. Rouce, Cliona M. Rooney, Gayatri Vyas, and Carlos A. Ramos
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,medicine.medical_treatment ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Immunotherapy, Adoptive ,Transplantation, Autologous ,CD19 ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,In vivo ,Positron Emission Tomography Computed Tomography ,Drug Discovery ,Genetics ,medicine ,Humans ,Molecular Biology ,B cell ,Aged ,Pharmacology ,Receptors, Chimeric Antigen ,biology ,business.industry ,Lymphoma, Non-Hodgkin ,Hematopoietic Stem Cell Transplantation ,CD28 ,hemic and immune systems ,Immunotherapy ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Chimeric antigen receptor ,Lymphoma ,Cytokine release syndrome ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Molecular Medicine ,Original Article ,Female ,business - Abstract
Second-generation (2G) chimeric antigen receptors (CARs) targeting CD19 are highly active against B cell malignancies, but it is unknown whether any of the costimulatory domains incorporated in the CAR have superior activity to others. Because CD28 and 4-1BB signaling activate different pathways, combining them in a single third-generation (3G) CAR may overcome the limitations of each individual costimulatory domain. We designed a clinical trial in which two autologous CD19-specific CAR-transduced T cell products (CD19.CARTs), 2G (with CD28 only) and 3G (CD28 and 4-1BB), were infused simultaneously in 16 patients with relapsed or refractory non-Hodgkin's lymphoma. 3G CD19.CARTs had superior expansion and longer persistence than 2G CD19.CARTs. This difference was most striking in the five patients with low disease burden and few circulating normal B cells, in whom 2G CD19.CARTs had limited expansion and persistence and correspondingly reduced area under the curve. Of the 11 patients with measurable disease, three achieved complete responses and three had partial responses. Cytokine release syndrome occurred in six patients but was mild, and no patient required anti-IL-6 therapy. Hence, 3G CD19.CARTs combining 4-1BB with CD28 produce superior CART expansion and may be of particular value when treating low disease burden in patients whose normal B cells are depleted by prior therapy.
- Published
- 2018
18. T-Cell Therapy for Lymphoma Using Nonengineered Multiantigen-Targeted T Cells Is Safe and Produces Durable Clinical Effects
- Author
-
Carlos A. Ramos, Ifigeneia Tzannou, Tao Wang, Rayne H. Rouce, George Carrum, Mrinalini Bilgi, Helen E. Heslop, Wendy L Callejas, Manik Kuvalekar, Ann M. Leen, Cliona M. Rooney, Mengfen J Wu, Adrian P. Gee, Catherine M. Bollard, Zihua Zeng, Bambi Grilley, Malcolm K. Brenner, Rammurti T. Kamble, Ayumi Watanabe, Premal Lulla, Juan F. Vera, and Spyridoula Vasileiou
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,Lymphoma ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Cell- and Tissue-Based Therapy ,MEDLINE ,Young Adult ,Antigens, Neoplasm ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Aged ,Salvage Therapy ,Chemotherapy ,Extramural ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,Prognosis ,medicine.disease ,medicine.anatomical_structure ,Female ,business - Abstract
PURPOSE Patients with relapsed lymphomas often fail salvage therapies including high-dose chemotherapy and mono-antigen–specific T-cell therapies, highlighting the need for nontoxic, novel treatments. To that end, we clinically tested an autologous T-cell product that targets multiple tumor-associated antigens (TAAs) expressed by lymphomas with the intent of treating disease and preventing immune escape. PATIENTS AND METHODS We expanded polyclonal T cells reactive to five TAAs: PRAME, SSX2, MAGEA4, SURVIVIN, and NY-ESO-1. Products were administered to 32 patients with Hodgkin lymphomas (n = 14) or non-Hodgkin lymphomas (n = 18) in a two-part phase I clinical trial, where the objective of the first phase was to establish the safety of targeting all five TAAs (fixed dose, 0.5 × 107 cells/m2) simultaneously and the second stage was to establish the maximum tolerated dose. Patients had received a median of three prior lines of therapy and either were at high risk for relapse (adjuvant arm, n = 17) or had chemorefractory disease (n = 15) at enrollment. RESULTS Infusions were safe with no dose-limiting toxicities observed in either the antigen- or dose-escalation phases. Although the maximum tolerated dose was not reached, the maximum tested dose at which efficacy was observed (two infusions, 2 × 107 cells/m2) was determined as the recommended phase II dose. Of the patients with chemorefractory lymphomas, two (of seven) with Hodgkin lymphomas and four (of eight) with non-Hodgkin lymphomas achieved durable complete remissions (> 3 years). CONCLUSION T cells targeting five TAAs and administered at doses of up to two infusions of 2 × 107 cells/m2 are well-tolerated by patients with lymphoma both as adjuvant and to treat chemorefractory lymphoma. Preliminary indicators of antilymphoma activity were seen in the chemorefractory cohort across both antigen- and dose-escalation phases.
- Published
- 2021
19. Allogeneic NKT Cells Expressing a CD19-Specific CAR in Patients with Relapsed or Refractory B-Cell Malignancies
- Author
-
Carlos A. Ramos, Amy N. Courtney, Simon N. Robinson, Olga Dakhova, Premal D. Lulla, Rammurti T. Kamble, George Carrum, Tao Wang, Chunchao Zhang, Erica J. Di Pierro, and Leonid S. Metelitsa
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
20. Donor-Derived Adoptive T-Cell Therapy Targeting Multiple Tumor Associated Antigens to Prevent Post-Transplant Relapse in Patients with ALL
- Author
-
Swati Naik, Spyridoula Vasileiou, Ifigeneia Tzannou, Manik Kuvalekar, Ayumi Watanabe, Catherine Robertson, Adrian P. Gee, Bambi Grilley, George Carrum, Rammurti T. Kamble, LaQuisa Hill, Robert A. Krance, Caridad Martinez, Bilal Omer, Stephen Gottschalk, Helen E. Heslop, Cliona M. Rooney, Juan F. Vera, Ann M. Leen, and Premal D. Lulla
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
21. The safety and clinical effects of administering a multiantigen-targeted T cell therapy to patients with multiple myeloma
- Author
-
Manik Kuvalekar, Adrian P. Gee, Juan F. Vera, Meng-Fen Wu, Rammurti T. Kamble, Tao Wang, Mrinalini Bilgi, Carlos A. Ramos, Ayumi Watanabe, Premal Lulla, Mira Jeong, Shamika Ketkar, Matthew French-Kim, Spyridoula Vasileiou, Betty Chung, George Carrum, Bambi Grilley, Ann M. Leen, Malcolm K. Brenner, Shivani Mukhi, Linghua Wang, Helen E. Heslop, Yumei Li, and Ifigeneia Tzannou
- Subjects
Oncology ,medicine.medical_specialty ,PRAME ,business.industry ,T cell ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,General Medicine ,medicine.disease ,Prior Therapy ,medicine.anatomical_structure ,Refractory ,Antigen ,Antigens, Neoplasm ,Internal medicine ,Concomitant ,medicine ,Humans ,Neoplasm Recurrence, Local ,Multiple Myeloma ,business ,Multiple myeloma ,Ex vivo - Abstract
Multiple myeloma (MM) is an almost always incurable malignancy of plasma cells. Despite the advent of new therapies, most patients eventually relapse or become treatment-refractory. Consequently, therapies with nonoverlapping mechanisms of action that are nontoxic and provide long-term benefit to patients with MM are greatly needed. To this end, we clinically tested an autologous multitumor-associated antigen (mTAA)-specific T cell product for the treatment of patients with high-risk, relapsed or refractory MM. In this study, we expanded polyclonal T cells from 23 patients with MM. T cells whose native T cell receptors were reactive toward five myeloma-expressed target TAAs (PRAME, SSX2, MAGEA4, Survivin, and NY-ESO-1) were enriched ex vivo. To date, we have administered escalating doses of these nonengineered mTAA-specific T cells (0.5 × 107 to 2 × 107 cells/m2) to 21 patients with MM, 9 of whom were at high risk of relapse after a median of 3 lines of prior therapy and 12 with active, relapsed or refractory disease after a median of 3.5 prior lines. The cells were well tolerated, with only two transient, grade III infusion-related adverse events. Furthermore, patients with active relapsed or refractory myeloma enjoyed a longer than expected progression-free survival and responders included three patients who achieved objective responses concomitant with detection of functional TAA-reactive T cell clonotypes derived from the infused mTAA product.
- Published
- 2020
22. High Incidence of Autoimmune Disease after Hematopoietic Stem Cell Transplantation for Chronic Granulomatous Disease
- Author
-
Robert A. Krance, Caridad Martinez, Swati Naik, Bilal Omer, Feliz O. Seeborg, Hao Liu, Meenakshi Hegde, Imelda C. Hanson, Malcolm K. Brenner, Nabil Ahmed, Kathryn Leung, Carl E. Allen, Meng-Fen Wu, Jordan S. Orange, Ghadir Sasa, Yassine Khaled, Sarah K. Nicholas, William T. Shearer, Asaf D. Yanir, George Carrum, Nicholas L. Rider, Stephen Gottschalk, Ivan K. Chinn, Helen E. Heslop, Lenora M. Noroski, and Lisa R. Forbes
- Subjects
medicine.medical_specialty ,Pancytopenia ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Disease ,Granulomatous Disease, Chronic ,Guillain-Barre Syndrome ,Chimerism ,Autoimmune Diseases ,03 medical and health sciences ,0302 clinical medicine ,Chronic granulomatous disease ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Adverse effect ,Alemtuzumab ,Autoimmune disease ,Transplantation ,Cytopenia ,business.industry ,Incidence ,Incidence (epidemiology) ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Unrelated Donors ,business ,Follow-Up Studies ,030215 immunology ,medicine.drug - Abstract
There is a lack of consensus regarding the role and method of hematopoietic stem cell transplantation (HSCT) on patients with chronic granulomatous disease (CGD). Long-term follow-up after HSCT in these patient population is essential to know its potential complications and decide who will benefit the most from HSCT. We report the outcome of HSCT and long-term follow-up in 24 patients with CGD, transplanted in our center from either related (n = 6) or unrelated (n = 18) donors, over a 12-year period (2003 to 2015), using high-dose alemtuzumab in the preparative regimen. We evaluated the incidence and timing of adverse events and potential risk factors. We described in detailed the novel finding of increased autoimmunity after HSCT in patients with CGD. At a median follow-up of 1460 days, 22 patients were full donor chimeras, and 2 patients had stable mixed chimerism. All assessable patients showed normalization of their neutrophil oxidative burst test. None of the patients developed grades II to IV acute graft-versus-host disease, and no patient had chronic graft-versus-host disease. Twelve of 24 patients developed 17 autoimmune diseases (ADs). Severe ADs (cytopenia and neuropathy) occurred exclusively in the unrelated donor setting and mainly in the first year after HSCT, whereas thyroid AD occurred in the related donor setting as well and more than 3 years after HSCT. Two patients died due to infectious complications after developing autoimmune cytopenias. One additional patient suffered severe brain injury. The remaining 21 patients have long-term Lansky scores ≥ 80. The outcome of HSCT from unrelated donors is comparable with related donors but might carry an increased risk of developing severe AD. A lower dose of alemtuzumab may reduce this risk and should be tested in further studies.
- Published
- 2018
23. Tumor-Specific T-Cells Engineered to Overcome Tumor Immune Evasion Induce Clinical Responses in Patients With Relapsed Hodgkin Lymphoma
- Author
-
Tamara Tripic, Malcolm K. Brenner, Cecilia Barese, Daniel Y. Lee, Hao Liu, Adrian P. Gee, Helen E. Heslop, Vicky Torrano, Catherine M. Bollard, Stephen Gottschalk, Olga Dakhova, Owen A. O'Connor, Youli Zu, Cliona M. Rooney, Gianpietro Dotti, Conrad Russell Y. Cruz, Andrea N. Marcogliese, Meng-Fen Wu, George Carrum, and Carlos A. Ramos
- Subjects
Adult ,Male ,0301 basic medicine ,Herpesvirus 4, Human ,Cancer Research ,Time Factors ,T-Lymphocytes ,medicine.medical_treatment ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Viral Matrix Proteins ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Recurrence ,Biology of Neoplasia ,Neoplasms ,Tumor Microenvironment ,medicine ,Humans ,Receptor ,Cells, Cultured ,Cell Proliferation ,Chemotherapy ,Tumor microenvironment ,business.industry ,Growth factor ,Receptor, Transforming Growth Factor-beta Type II ,Genetic Therapy ,Immunotherapy ,Middle Aged ,Hodgkin Disease ,Phenotype ,Treatment Outcome ,030104 developmental biology ,Oncology ,Tumor Escape ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business - Abstract
Purpose Transforming growth factor-β (TGF-β) production in the tumor microenvironment is a potent and ubiquitous tumor immune evasion mechanism that inhibits the expansion and function of tumor-directed responses; therefore, we conducted a clinical study to discover the effects of the forced expression of a dominant-negative TGF-β receptor type 2 (DNRII) on the safety, survival, and activity of infused tumor-directed T cells. Materials and Methods In a dose escalation study, eight patients with Epstein Barr virus–positive Hodgkin lymphoma received two to 12 doses of between 2 × 107 and 1.5 × 108 cells/m2 of DNRII-expressing T cells with specificity for the Epstein Barr virus–derived tumor antigens, latent membrane protein (LMP)-1 and LMP-2 (DNRII-LSTs). Lymphodepleting chemotherapy was not used before infusion. Results DNRII-LSTs were resistant to otherwise inhibitory concentrations of TGF-β in vitro and retained their tumor antigen–specific activity. After infusion, the signal from transgenic T cells in peripheral blood increased up to 100-fold as measured by quantitative polymerase chain reaction for the transgene, with a corresponding increase in the frequency of functional LMP-specific T cells. Expansion was not associated with any acute or long-term toxicity. DNRII-LSTs persisted for up to ≥ 4 years. Four of the seven evaluable patients with active disease achieved clinical responses that were complete and ongoing in two patients at > 4 years, including in one patient who achieved only a partial response to unmodified tumor-directed T cells. Conclusion TGF-β–resistant tumor-specific T cells safely expand and persist in patients with Hodgkin lymphoma without lymphodepleting chemotherapy before infusion. DNRII-LSTs can induce complete responses even in patients with resistant disease. Expression of DNRII may be useful for the many other tumors that exploit this potent immune evasion mechanism.
- Published
- 2018
24. 37. Allogeneic, Off-the-Shelf, SARS-CoV-2-specific T Cells Demonstrate Reactivity Against Emerging Variant Strains
- Author
-
Spyridoula Vasileiou, Manik Kuvalekar, Aster Workineh, Ayumi Watanabe, Yovana Velazquez, Suhasini Lulla, Helen Elisabeth Heslop, Kimberly Mooney, Kevin Grimes, George Carrum, LaQuisa Hill, Premal Lulla, and Ann Marie Leen
- Subjects
Infectious Diseases ,AcademicSubjects/MED00290 ,Oncology ,Oral Abstracts - Abstract
Background The impact of COVID-19 has been profound with >170,000,000 confirmed cases worldwide and emerging variants being a cause of global concern. Defects in T-cell function and trafficking have been described among those with severe illness, and immunodeficiency is a risk factor for persistent viral shedding and prolonged symptoms. Because of our prior clinical data demonstrating that allogeneic, off-the-shelf virus-specific T cells (VSTs) can safely and effectively treat viral infections, we investigated the feasibility of targeting COVID-19 using banked, SARS-CoV-2-specific VSTs. Methods We first screened PBMCs from convalescent individuals against 18 structural and non-structural/accessory (NSPs/APs) SARS-CoV-2 proteins and identified 5 [Spike (S), Membrane (M), Nucleoprotein (N), NSP4, and AP7a] as immunodominant which were then advanced to our VST production process. Results Using overlapping peptide libraries spanning these antigens as a stimulus, we achieved a mean 7.6±0.9 fold expansion (n=13) of VSTs (96±0.5%), with a mixture of cytotoxic (CD8+) and helper (CD4+) T cells that expressed activation and central/effector memory markers. These VSTs were potent, Th1-polarized and polyfunctional, producing IFNγ, TNFα, GM-CSF and Granzyme B. Moreover, the VSTs were able to kill pepmix-loaded autologous targets with no evidence of auto- or alloreactivity, attesting to their virus selectivity and safety for clinical use (Figure 1). Finally, though initially generated against the reference strain NC_045512.2 (Wuhan), these VSTs were able to recognize other clinically important variants including B1.1.7 (UK), B1.351 (South Africa) and P1 (Brazil). This demonstrates the cross-reactive potential of these polyclonal and diverse VSTs, which were developed to provide potent antiviral effects and minimize the risk of immune escape due to sequence variation. Figure 1: SARS-CoV-2 Specific T cells Have Demonstrated Selective Cytolytic Activity against SARS-CoV-2 While Leaving Non-Virus Infected Targets Intact. Conclusion In conclusion, it is feasible to generate polyclonal SARS-CoV-2 VSTs that provide coverage against variant strains using GMP-compliant manufacturing methodologies. We have advanced this product to the bedside for administration in a Phase I, randomized clinical trial [VSTs+ standard of care (SOC) vs SOC] in high-risk patients hospitalized with COVID-19 (NCT04401410). Disclosures Spyridoula Vasileiou, PhD, AlloVir (Consultant) Manik Kuvalekar, MSc, AlloVir (Consultant) Aster Workineh, MSc, AlloVir (Employee) Ayumi Watanabe, BSc, AlloVir (Consultant) Yovana Velazquez, BSc, AlloVir (Consultant) Helen Elisabeth Heslop, MD, AlloVir (Shareholder)Cell Medica (Grant/Research Support)Gilead Sciences (Consultant)Kiadis Pharma (Consultant)Marker Therapeutics (Consultant, Shareholder)Mesoblast (Consultant)Novartis (Consultant)PACT Pharma (Consultant)Tessa Therapeutics (Consultant, Research Grant or Support) Premal Lulla, MD, Johnson & Johnson (Shareholder) Ann Marie Leen, PhD, AlloVIr (Consultant, Shareholder)Marker Therapeutics (Consultant, Shareholder)
- Published
- 2021
25. Excellent Outcomes for Pediatric and Adult Patients with Hodgkin Lymphoma Receiving Rituximab As Adjunctive Therapy to BEAM Conditioning for Autologous Stem Cell Transplantation
- Author
-
Baheyeldin Salem, LaQuisa Hill, Shreeya Patel, Tami John, Caridad Martinez, Brian D. Friend, George Carrum, John Craddock, Robert A. Krance, Premal Lulla, Rammurti T. Kamble, Carlos A. Ramos, Helen E. Heslop, Ghadir S. Sasa, Erin E Doherty, Khaled Yassine, and Ibrahim N. Muhsen
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Adult patients ,business.industry ,Cell Biology ,Hematology ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Molecular Medicine ,Immunology and Allergy ,Hodgkin lymphoma ,Rituximab ,business ,medicine.drug - Published
- 2021
26. Allogeneic, Off-the-Shelf, Sars-Cov-2-Specific T Cells to Treat High-Risk Patients with COVID-19
- Author
-
Kevin Grimes, Ayumi Watanabe, Helen E. Heslop, Premal Lulla, Yovana Velazquez, Aster Workineh, Manik Kuvalekar, Spyridoula Vasileiou, Ann M. Leen, Kimberly Mooney, Suhasini Lulla, LaQuisa Hill, and George Carrum
- Subjects
Transplantation ,High risk patients ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Cell Biology ,Hematology ,Poster Session - Infectious Diseases and Cytotoxic T Lymphocytes ,Virology ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Off the shelf ,business - Published
- 2021
27. Allogeneic NKT Cells Expressing a CD19-Specific CAR in Patients with Relapsed or Refractory B-Cell Malignancies: An Interim Analysis
- Author
-
Erica J Di Pierro, Leonid S. Metelitsa, Rammurti T. Kamble, Tao Wang, Simon N. Robinson, Amy N. Courtney, Olga Dakhova, Premal Lulla, George Carrum, Carlos A. Ramos, and Chunchao Zhang
- Subjects
biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,Interim analysis ,Natural killer T cell ,Biochemistry ,CD19 ,medicine.anatomical_structure ,Refractory ,Cancer research ,biology.protein ,Medicine ,In patient ,business ,B cell - Abstract
Autologous T cells engineered to express a CD19-specific chimeric antigen receptor (CAR) mediate high rates of complete response (CR) in patients with B-cell malignancies. However, autologous cell therapy products are time- and resource-intensive to manufacture and vary in potency and toxicity. Unlike polymorphic HLA-restricted T cells, monomorphic CD1d-restricted Vα24-invariant natural killer T cells (NKTs) are not alloreactive, and therefore therapeutic NKTs can be generated from allogeneic donors without the risk of graft-versus-host disease (GvHD). Moreover, pre-clinical models suggest that CAR-NKTs have inherent advantages over CAR-T products including an ability to trans-activate NK cells, cross-prime tumor-specific CD8 T cells, and recognize CD1d-positive B-cell lymphoma cells via their endogenous NKT T cell receptor. We report interim results from five patients treated on a phase 1 dose-escalation trial of allogeneic NKTs engineered to co-express a CD19-specific CAR, IL-15, and shRNA targeting beta-2 microglobulin and CD74 for downregulation of HLA class I and class II molecules, respectively (ANCHOR, NCT00840853). Four patients with relapsed/refractory B-cell non-Hodgkin's lymphoma (NHL, cohort A) were enrolled on dose level (DL) 1 (NHL-1, -2, -3) and DL 2 (NHL-4), and 1 patient with relapsed acute B-lymphoblastic leukemia (ALL, cohort B) was enrolled on DL 1 (ALL-1). Primary and secondary objectives of the trial are to assess safety and anti-tumor responses; immune response evaluation is an additional objective. NKTs were isolated from the leukapheresis product of 1 HLA-unmatched healthy individual, transduced with the CAR, expanded ex vivo for 14 days to a total of 2.7×10 9 CAR-positive cells (99.8% NKT purity, 0.04% T cells), and cryopreserved. Patients received 10 7 (DL 1) or 3×10 7 (DL 2) CAR-NKT cells per square meter of body surface area following lymphodepleting conditioning with cyclophosphamide/fludarabine. Adverse events were evaluated per NCI criteria. When accessible, patients underwent core biopsies of an involved site at 2-5 weeks post-infusion. Response to therapy was assessed at 4 weeks per Lugano Criteria (for NHL) or NCCN guidelines (for ALL). The most common adverse effects observed were nausea and grade 3-4 hematologic toxicities related to the lymphodepletion chemotherapy. There were no early adverse events attributable to the cellular product except grade 1 cytokine release syndrome in 1 patient. Of the 4 NHL patients (all with diffuse large B cell lymphoma), 3 had a partial response (NHL-1, -2, and -4, Figure 1A) that evolved into a CR in 1 case (NHL-2). The ALL patient achieved a CR with incomplete hematologic recovery and showed no evidence of leukemia by morphology, flow cytometry, or next-generation sequencing at 4 weeks. We detected in vivo expansion of donor-derived NKT and CAR-NKT cells in the peripheral blood of NHL-4 and ALL-1 that peaked at 1 week post-infusion in both cases as determined by flow cytometry and qPCR. While CAR-NKTs were not detected in the peripheral blood of the first 3 NHL patients beyond 3 hours post-infusion, they were found in tumor tissues collected from the 2 biopsied NHL patients (Figure 1B). In patient NHL-2, we also observed a 2000-fold expansion of recipient NKTs with a skewed TCRβ repertoire; this population peaked at 6 weeks post-treatment and remained elevated through 12 weeks (Figure 1C). An ELISpot assay performed with peripheral blood from NHL-1 and NHL-2 before treatment and at 4 and 6 weeks post-therapy to detect changes in the frequency of T cells reactive to a set of tumor-associated antigens (TAAs) found no evidence for epitope spreading toward the tested TAAs. In conclusion, our data indicate that allogeneic CAR-NKT cells are well-tolerated and can mediate objective responses in relapsed/refractory NHL and ALL patients even at the low doses tested. Our initial results from this first-in-human clinical evaluation of allogeneic CAR-NKTs suggest that NKTs represent a promising platform for "off-the-shelf" cancer immunotherapy. Figure 1 Figure 1. Disclosures Ramos: Tessa Therapeutics: Patents & Royalties, Research Funding; Athenex: Research Funding; Genentech: Consultancy; Novartis: Consultancy. Courtney: Athenex: Patents & Royalties, Research Funding. Metelitsa: Athenex: Patents & Royalties, Research Funding. OffLabel Disclosure: Cyclophosphamide and fludarabine are being used as lymphodepleting agents before immune effector infusion.
- Published
- 2021
28. Donor-Derived Adoptive T-Cell Therapy Targeting Multiple Tumor Associated Antigens to Prevent Post-Transplant Relapse in Patients with ALL
- Author
-
Bilal Omer, Catherine Robertson, Spyridoula Vasileiou, LaQuisa Hill, Juan F. Vera, Adrian P. Gee, Caridad Martinez, Ifigeneia Tzannou, George Carrum, Cliona M. Rooney, Helen E. Heslop, Bambi Grilley, Swati Naik, Stephen Gottschalk, Robert A. Krance, Rammurti T. Kamble, Ann M. Leen, Ayumi Watanabe, Premal Lulla, and Manik Kuvalekar
- Subjects
business.industry ,T cell ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Post transplant ,medicine.anatomical_structure ,Antigen ,Cancer research ,Medicine ,In patient ,Donor derived ,Multiple tumors ,business - Abstract
Background: Hematopoietic stem cell transplant (HSCT) is a curative option for patients with high-risk Acute Lymphoblastic Leukemia (HR-ALL), but relapse remains a major cause of treatment failure. Strategies to enhance the graft-versus-leukemia (GVL) effect have been employed to prevent relapse, including modulating immune suppression post-HSCT to hasten immune reconstitution or with the use of donor lymphocyte infusions (DLIs). However, DLIs carry a significant risk of graft-versus-host disease (GVHD) due to the concurrent transfer of alloreactive T cells. To enhance the GVL effect while minimizing GVHD, we developed a protocol for the generation of ex vivo expanded, donor-derived T-cell lines targeting PRAME, WT1 and Survivin - tumor associated antigens that are frequently expressed in both B- and T-cell ALL. These multi-antigen-targeted T cells (multiTAAs) were adoptively transferred to pediatric and adult patients with HR-ALL who had undergone an allogeneic HSCT. Methods: Donor-derived multiTAA-specific T cells were generated by co-culturing PBMCs with autologous DCs loaded with pepmixes (15 mer peptides overlapping by 11 amino acids) spanning all 3 target antigens in the presence of a Th1-polarizing/pro-proliferative cytokine cocktail. Following 2-4 rounds of stimulation these multiTAA-specific T cells were infused to patients with ALL who had undergone an HSCT but remained at a high risk for disease relapse. Results: We have generated 15 clinical grade multiTAA-specific T cell lines comprising CD3+ T cells (mean 95.1±1.9%) with a mixture of CD4+ (mean 22.8±6.3%) and CD8+ (mean 52.5±5.3%) cells, which expressed central [CD45RO+/CD62L+: 13.5±2.8%] and effector memory markers [CD45RO+/CD62L-: 56.4±3.8%]. The expanded lines recognized the targeted antigens PRAME (range 0-370 SFC/2x10 5), WT1 (0-363 SFC/2x10 5), and Survivin (0-65 SFC/2x10 5) in an IFNg ELIspot. None of the lines reacted against non-malignant patient-derived cells (3.7±0.8% specific lysis; E: T 20:1) - a study release criterion indicating lack of alloreactivity. We have infused 11 HR-ALL patients (8 pediatric and 3 adult) with donor-derived multiTAA-specific T cells to prevent disease relapse (Table 1). Patients were administered with up to 4 infusions of cells at 3 escalating dose levels, ranging from 0.5 - 2x10 7 cells/m 2. Infusions were well tolerated with no dose-limiting toxicity, GVHD, cytokine release syndrome or other adverse events. Three patients were not evaluable per study criteria as they received >0.5mg/kg of steroids (2 patients received stress doses for septic shock and 1 for elevated liver enzymes presumed to be GVHD that was later ruled out) within 4 weeks of infusion and were replaced. Six of the 8 remaining patients infused remain in CR on long-term follow up at a median of 46.5 months post-infusion (range 9-51 months). In patients who remained in long term CR we detected an expansion of tumor-reactive T cells in their peripheral blood post-infusion against both targeted (WT1, Survivin, PRAME) and non-targeted antigens (SSX2, MAGE-A4, -A1, -A2B, -C1, MART1, AFP and NYESO1) reflecting epitope and antigen spreading, which correlated temporally (within 4 weeks) with multiTAA infusions. By contrast in the two patients who relapsed we saw no evidence of in vivo T cell amplification within the first 4 weeks after infusion. Conclusion: The preparation and infusion of donor-derived multiTAA-specific T cells to patients with B- and T-ALL post allogeneic HSCT is feasible, safe and as evidenced by in vivo tumor-directed T cell expansion and antigen spreading in patients, may contribute to disease control. This strategy may present a promising addition to current immunotherapeutic approaches for prophylaxis for leukemic relapse in HSCT recipients. Figure 1 Figure 1. Disclosures Vasileiou: Allovir: Consultancy. Tzannou: Gileas: Honoraria; Allovir: Current equity holder in publicly-traded company. Kuvalekar: Allovir: Consultancy. Watanabe: Allovir: Consultancy. Grilley: QB Regulatory Consulting: Other: Ownership, project management support, Research Funding; Marker: Consultancy, Other: Regulatory and project management support; Allovir: Current equity holder in publicly-traded company, Other: Leadership. Hill: Incyte: Membership on an entity's Board of Directors or advisory committees. Omer: Allovir: Research Funding. Gottschalk: Tessa Therapeutics: Consultancy; Immatics: Membership on an entity's Board of Directors or advisory committees; Other: Other: patents and patent applications in the field of cancer cell and gene therapy ; Tidal: Consultancy; Novartis: Consultancy; Catamaran Bio: Consultancy. Heslop: Gilead: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Kiadis: Membership on an entity's Board of Directors or advisory committees; Kuur Therapeutics: Research Funding; GSK: Membership on an entity's Board of Directors or advisory committees; Allovir: Current equity holder in publicly-traded company; Tessa Therapeutics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Marker Therapeutics: Current equity holder in publicly-traded company; Fresh Wind Biotherapies: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees. Rooney: Allogene: Patents & Royalties; Bellicum: Patents & Royalties; Bluebird: Current equity holder in publicly-traded company; Allovir: Current equity holder in publicly-traded company; Alimera: Consultancy; Memgen: Consultancy; TScan Therapeutics: Consultancy; Takeda: Patents & Royalties; Marker: Current equity holder in publicly-traded company; Tessa: Consultancy, Other: Leadership, Research Funding. Vera: Allovir: Consultancy, Current equity holder in publicly-traded company, Other: Leadership, travel , accomodations, expenses, Patents & Royalties; Marker: Current Employment, Other: Travel, Accomodations, Expenses, Patents & Royalties, Research Funding. Leen: Allovir: Consultancy, Current equity holder in publicly-traded company; Marker: Current equity holder in publicly-traded company.
- Published
- 2021
29. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation
- Author
-
Bilal Omer, Premal Lulla, Bambi J. Grilley, Helen E. Heslop, Cliona M. Rooney, Carlos A. Ramos, Swati Naik, Ayumi Watanabe, Kathryn Leung, Adrian P. Gee, Manik Kuvalekar, Ifigeneia Tzannou, Hao Liu, Caridad Martinez, Stephen Gottschalk, George Carrum, Robert A. Krance, Ann M. Leen, Anastasia Papadopoulou, Meng Fen Wu, Malcolm K. Brenner, and Ghadir Sasa
- Subjects
Adult ,Male ,0301 basic medicine ,Herpesvirus 4, Human ,Cancer Research ,Herpesvirus 6, Human ,T-Lymphocytes ,viruses ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,medicine.disease_cause ,Immunotherapy, Adoptive ,Virus ,Epitope ,03 medical and health sciences ,medicine ,Humans ,Transplantation, Homologous ,biology ,business.industry ,Adenoviruses, Human ,DNA Viruses ,Hematopoietic Stem Cell Transplantation ,ORIGINAL REPORTS ,Immunotherapy ,biology.organism_classification ,medicine.disease ,Virology ,DNA Virus Infections ,BK virus ,Transplantation ,Treatment Outcome ,030104 developmental biology ,Oncology ,BK Virus ,Immunology ,Female ,Human herpesvirus 6 ,business ,Hemorrhagic cystitis - Abstract
Purpose Improvement of cure rates for patients treated with allogeneic hematopoietic stem-cell transplantation (HSCT) will require efforts to decrease treatment-related mortality from severe viral infections. Adoptively transferred virus-specific T cells (VSTs) generated from eligible, third-party donors could provide broad antiviral protection to recipients of HSCT as an immediately available off-the-shelf product. Patient and Methods We generated a bank of VSTs that recognized five common viral pathogens: Epstein-Barr virus (EBV), adenovirus (AdV), cytomegalovirus (CMV), BK virus (BKV), and human herpesvirus 6 (HHV-6). The VSTs were administered to 38 patients with 45 infections in a phase II clinical trial. Results A single infusion produced a cumulative complete or partial response rate of 92% (95% CI, 78.1% to 98.3%) overall and the following rates by virus: 100% for BKV (n = 16), 94% for CMV (n = 17), 71% for AdV (n = 7), 100% for EBV (n = 2), and 67% for HHV-6 (n = 3). Clinical benefit was achieved in 31 patients treated for one infection and in seven patients treated for multiple coincident infections. Thirteen of 14 patients treated for BKV-associated hemorrhagic cystitis experienced complete resolution of gross hematuria by week 6. Infusions were safe, and only two occurrences of de novo graft-versus host disease (grade 1) were observed. VST tracking by epitope profiling revealed persistence of functional VSTs of third-party origin for up to 12 weeks. Conclusion The use of banked VSTs is a feasible, safe, and effective approach to treat severe and drug-refractory infections after HSCT, including infections from two viruses (BKV and HHV-6) that had never been targeted previously with an off-the-shelf product. Furthermore, the multispecificity of the VSTs ensures extensive antiviral coverage, which facilitates the treatment of patients with multiple infections.
- Published
- 2017
30. Impact of Routine Surveillance Imaging on Outcomes of Patients With Diffuse Large B-Cell Lymphoma After Autologous Hematopoietic Cell Transplantation
- Author
-
Mehdi Hamadani, Narendranath Epperla, George Carrum, Timothy S. Fenske, Sai Ravi Pingali, Reem Karmali, Jonathan Kapke, Asmita Patel, Parameswaran Hari, Kristin Richardson, Namrata Shah, and Sravanthi P. Teegavarapu
- Subjects
Diagnostic Imaging ,Male ,Cancer Research ,medicine.medical_specialty ,Time Factors ,Radiography ,Salvage therapy ,Transplantation, Autologous ,Gastroenterology ,Workflow ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Recurrence ,immune system diseases ,Positron Emission Tomography Computed Tomography ,hemic and lymphatic diseases ,Internal medicine ,Humans ,Medicine ,Monitoring, Physiologic ,Neoplasm Staging ,Retrospective Studies ,Postoperative Care ,business.industry ,Hematopoietic Stem Cell Transplantation ,Retrospective cohort study ,Hematology ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Lymphoma ,Surgery ,Transplantation ,Treatment Outcome ,surgical procedures, operative ,Oncology ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Female ,Lymphoma, Large B-Cell, Diffuse ,Surveillance imaging ,Tomography, X-Ray Computed ,business ,Diffuse large B-cell lymphoma ,030215 immunology - Abstract
Background For patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL), autologous hematopoietic cell transplantation (auto-HCT) is commonly used. After auto-HCT, DLBCL patients are often monitored with surveillance imaging. However, there is little evidence to support this practice. Patients and Methods We performed a multicenter retrospective study of DLBCL patients who underwent auto-HCT (n = 160), who experienced complete remission after transplantation, and who then underwent surveillance imaging. Of these, only 45 patients experienced relapse after day +100 after auto-HCT, with relapse detected by routine imaging in 32 (71%) and relapse detected clinically in 13 (29%). Results Baseline patient characteristics were similar between the 2 groups. Comparing the radiographic and clinically detected relapse groups, the median time from diagnosis to auto-HCT (389 days vs. 621 days, P = .06) and the median follow-up after auto-HCT (2464 days vs. 1593 days P = .60) were similar. The median time to relapse after auto-HCT was 191 days in radiographically detected relapses compared to 492 days in clinically detected relapses ( P = .35), and median postrelapse survival was 359 days in such patients compared to 123 days in patients with clinically detected relapse ( P = .36). However, the median posttransplantation overall survival was not significantly different for patients with relapse detected by routine imaging versus relapse detected clinically (643 vs. 586 days, P = .68). Conclusion A majority (71%) of DLBCL relapses after auto-HCT are detected by routine surveillance imaging. Overall, there appears to be limited utility for routine imaging after auto-HCT except in select cases where earlier detection and salvage therapy with allogeneic HCT is a potential option.
- Published
- 2016
31. Health disparities experienced by Black Americans with multiple myeloma in the United States: A population-based study
- Author
-
Deepa Dongarwar, Hamisu M. Salihu, LaQuisa Hill, Carlos A. Ramos, Saad Z. Usmani, Helen E. Heslop, Samer Al Hadidi, George Carrum, Premal Lulla, and Rammurti T. Kamble
- Subjects
Population based study ,Cancer Research ,Oncology ,business.industry ,Incidence (epidemiology) ,Mortality rate ,Hematologic malignancy ,Medicine ,business ,medicine.disease ,Health equity ,Multiple myeloma ,Demography - Abstract
e18512 Background: Multiple Myeloma (MM) is the most common hematologic malignancy in Black Americans. Incidence and death rates for MM in Black Americans are more than double those in Whites. Our study aimed to evaluate trends of all cause in-hospital mortality among Black Americans with MM and to investigate characteristics of MM-related hospitalizations. Methods: We conducted a retrospective cross-sectional study of hospitalizations in adult patients with MM during 2008-2017 using the National Inpatient Sample (NIS), the largest all-payer inpatient care database in the US. We used joinpoint regression to assess temporal trends in the national incidence of in-hospital death. We conducted adjusted survey logistic regression to generate adjusted odds ratios to measure the likelihood of in-hospital death among MM related hospitalizations. Results: Admissions related to MM constituted 0.32% of all hospitalizations in the study period (913,967 out of 285,876,821). The prevalence of MM related hospitalizations was higher in Black Americans when compared with Whites (476.0 vs 305.6 per 100,000 hospitalizations, p
- Published
- 2021
32. Efficacy of Letermovir for CMV Prophylaxis Following Allogeneic Hematopoietic Stem Cell Transplant T-Cell Depleted with Alemtuzumab
- Author
-
Helen E. Heslop, Rammurti T. Kamble, Tao Wang, George Carrum, Carlos A. Ramos, LaQuisa Hill, Premal Lulla, and Ibrahim N. Muhsen
- Subjects
Transplantation ,business.industry ,T cell ,Cell Biology ,Hematology ,Letermovir ,medicine.anatomical_structure ,Cmv prophylaxis ,Immunology ,medicine ,Molecular Medicine ,Immunology and Allergy ,Alemtuzumab ,Allogeneic hematopoietic stem cell transplant ,business ,medicine.drug - Published
- 2021
33. Clinical responses with T lymphocytes targeting malignancy-associated κ light chains
- Author
-
Enli Liu, Zhuyong Mei, Hao Liu, Rammurti T. Kamble, Bambi Grilley, Barbara Savoldo, Helen E. Heslop, Malcolm K. Brenner, Brandon Ballard, Huimin Zhang, Gianpietro Dotti, Olga Dakhova, Adrian P. Gee, George Carrum, Meng Fen Wu, Cliona M. Rooney, Vicky Torrano, and Carlos A. Ramos
- Subjects
Adult ,Male ,0301 basic medicine ,Adoptive cell transfer ,Cyclophosphamide ,T-Lymphocytes ,T cell ,Chronic lymphocytic leukemia ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Enzyme-Linked Immunosorbent Assay ,CD19 ,Immunophenotyping ,Immunoglobulin kappa-Chains ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,B cell ,Aged ,biology ,business.industry ,Lymphoma, Non-Hodgkin ,Remission Induction ,General Medicine ,Middle Aged ,medicine.disease ,Adoptive Transfer ,Leukemia, Lymphocytic, Chronic, B-Cell ,3. Good health ,Retroviridae ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Feasibility Studies ,Female ,Clinical Medicine ,business ,medicine.drug - Abstract
BACKGROUND. Treatment of B cell malignancies with adoptive transfer of T cells with a CD19-specific chimeric antigen receptor (CAR) shows remarkable clinical efficacy. However, long-term persistence of T cells targeting CD19, a pan–B cell marker, also depletes normal B cells and causes severe hypogammaglobulinemia. Here, we developed a strategy to target B cell malignancies more selectively by taking advantage of B cell light Ig chain restriction. We generated a CAR that is specific for the κ light chain (κ.CAR) and therefore recognizes κ-restricted cells and spares the normal B cells expressing the nontargeted λ light chain, thus potentially minimizing humoral immunity impairment. METHODS. We conducted a phase 1 clinical trial and treated 16 patients with relapsed or refractory κ+ non-Hodgkin lymphoma/chronic lymphocytic leukemia (NHL/CLL) or multiple myeloma (MM) with autologous T cells genetically modified to express κ.CAR (κ.CARTs). Other treatments were discontinued in 11 of the 16 patients at least 4 weeks prior to T cell infusion. Six patients without lymphopenia received 12.5 mg/kg cyclophosphamide 4 days before κ.CART infusion (0.2 × 108 to 2 × 108 κ.CARTs/m2). No other lymphodepletion was used. RESULTS. κ.CART expansion peaked 1–2 weeks after infusion, and cells remained detectable for more than 6 weeks. Of 9 patients with relapsed NHL or CLL, 2 entered complete remission after 2 and 3 infusions of κ.CARTs, and 1 had a partial response. Of 7 patients with MM, 4 had stable disease lasting 2–17 months. No toxicities attributable to κ.CARTs were observed. CONCLUSION. κ.CART infusion is feasible and safe and can lead to complete clinical responses. TRIAL REGISTRATION. ClinicalTrials.gov {"type":"clinical-trial","attrs":{"text":"NCT00881920","term_id":"NCT00881920"}}NCT00881920. FUNDING. National Cancer Institute (NCI) grants 3P50CA126752 and 5P30CA125123 and Leukemia and Lymphoma Society (LLS) Specialized Centers of Research (SCOR) grant 7018.
- Published
- 2016
34. Using Allogeneic, Off-the-Shelf, Sars-Cov-2-Specific T Cells to Treat High Risk Patients with COVID-19
- Author
-
George Carrum, Ann M. Leen, Kimberly Mooney, Kevin Grimes, Helen E. Heslop, Spyridoula Vasileiou, LaQuisa Hill, Aster Workineh, Ayumi Watanabe, Manik Kuvalekar, Premal Lulla, Yovana Velazquez, and Suhasini Lulla
- Subjects
business.industry ,ELISPOT ,T cell ,Immunology ,CD28 ,Cell Biology ,Hematology ,Biochemistry ,703.Adoptive Immunotherapy: Mechanisms and New Approaches ,Immune system ,medicine.anatomical_structure ,Antigen ,Cytotoxic T cell ,Medicine ,IL-2 receptor ,business ,CD8 - Abstract
Background. On 11 March, 2020 the World Health Organization declared COVID-19, caused by SARS-CoV-2, a global pandemic with almost 17,000,000 confirmed cases worldwide by the end of July, of which 4,500,000 were in the US. Approximately 20% of patients develop severe disease that can evolve into acute respiratory distress syndrome leading to respiratory or multiorgan failure, with an overall mortality of up to 4%. Older age, comorbidities such as hypertension and diabetes, and immune compromise have been identified as major risk factors associated with poor prognosis. For example, in immunocompromised HSCT patients mortality rates as high as 20% have been reported (www.cibmtr.org/COVID19). Furthermore, there is accumulating evidence regarding the protective role of T cells, with reduced counts and dysregulation seen more prominently in individuals with severe rather than mild COVID-19. Our group has previously demonstrated the feasibility, safety and clinical efficacy of administering allogeneic ex vivo expanded multivirus-specific T cells (multi-VSTs) as a banked, off-the-shelf product for the treatment of EBV, CMV, BKV, HHV6 and AdV infections/disease in immunocompromised individuals. Given the lack of preventative or therapeutic agents and the emerging evidence of the pivotal protective role of SARS-CoV-2-specific CD4+ and CD8+ T cells, we sought to explore the feasibility of developing a banked, SARS-CoV-2-specific VST product to treat those at highest risk of severe COVID-19 disease (i.e. HSCT recipients, elderly individuals, patients with comorbidities). Methods. To first identify immunogenic and protective SARS-CoV-2 antigens we screened PBMCs from convalescent individuals with mild COVID-19 (not requiring hospitalization) for T cell activity against overlapping peptide libraries (pepmixes) spanning 18 structural and non-structural SARS-CoV-2 proteins of which 8 [structural proteins: Spike (S), Membrane (M) and Nucleoprotein (N); non-structural proteins (Nsps): 3, 4, 6, 12; and the accessory protein (AP) 7a] were identified as immunodominant and advanced for VST manufacturing. We subsequently utilized our optimized VST manufacturing process and culture in a G-Rex device in medium supplemented with activating cytokines to generate SARS-CoV-2-specific T cells with activity against this combination of immunodominant targets. Results. We achieved a mean 29±7 fold expansion (mean±SEM; n=5) of cells that were comprised almost exclusively of CD3+ T cells (97.1±0.7%; mean±SEM), with a mixture of cytotoxic (CD8+; 10.2±1.2%) and helper (CD4+; 85.5±1.8%) T cells. These cells had a phenotype consistent with effector function and memory potential, as evidenced by upregulation of the activation markers CD25, CD69, and CD28 and expression of central (CD45RO+/CD62L+) and effector memory markers (CD45RO+/CD62L−), with minimal PD1 or Tim3 expression. To confirm the anti-viral activity of our expanded cells we performed an IFNγ ELIspot using each of the individual stimulating antigens as an immunogen and all lines proved to be reactive against the target antigens [S: 2,118±479 SFC/2x105; M: 1,084±182; N: 1,124±335; Nsp3: 71±48.6; Nsp4: 68±30; Nsp6: 23±6.7; AP7a: 65±43; and Nsp12: 29±9]. As demonstrated by intracellular cytokine staining (ICS), the immune response was mediated by both CD4+ and CD8+ T cell subsets, and the majority of IFNg-producing cells also produced TNFa. Reactive cells exhibited a primarily Th1-polarized profile as measured by Granzyme B production, luminex array and single-cell protein analysis. In addition, the expanded cells were able to kill viral pepmix-loaded autologous PHA blasts with minimal/no activity against non-antigen-expressing autologous and allogeneic targets (Figure 1). Conclusion. SARS-CoV-2 VSTs generated from convalescent individuals are Th1-polarized, polyfunctional and selectively able to kill viral antigen-expressing targets with no auto- or alloreactivity, indicative of both their selectivity and safety for clinical use. We are rapidly advancing this product to the clinic for administration in a randomized clinical trial (VSTs+SOC vs SOC) to prevent the development of severe disease in high risk hospitalized patients such as those post-transplant or therapy for hematologic malignancy. Figure 1: SARS-CoV-2 Specific T cells Demonstrate Selective Cytolytic Activity against viral antigen-expressing targets. Figure 1 Disclosures Vasileiou: AlloVir: Consultancy. Kuvalekar:AlloVir: Consultancy. Workineh:AlloVir: Current Employment. Watanabe:AlloVir: Consultancy. Heslop:Novartis: Consultancy; Gilead Biosciences: Consultancy; PACT Pharma: Consultancy; Kiadis: Consultancy; Tessa Therapeutics: Consultancy, Research Funding; AlloVir: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Marker Therapeutics: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees. Hill:Incyte: Membership on an entity's Board of Directors or advisory committees. Leen:AlloVir: Consultancy, Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees; Marker Therapeutics: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2020
35. Assessment and reporting of quality-of-life measures in pivotal clinical trials of hematological malignancies
- Author
-
George Carrum, Carlos A. Ramos, Helen E. Heslop, Samer Al Hadidi, and Rammurti T. Kamble
- Subjects
Clinical trial ,Health related quality of life ,Cancer Research ,medicine.medical_specialty ,Quality of life (healthcare) ,Oncology ,business.industry ,medicine ,Intensive care medicine ,business ,humanities - Abstract
158 Background: The importance of assessment and reporting of health related quality of life (QoL) has been increasingly recognized in the field of oncology. QoL measures, which reflect patient’s perceived benefits and satisfaction, might be especially important in hematological malignancies clinical trials, where the intervention may not result in cure with modest overall survival benefit. Methods: Data on health related QoL were obtained from studies’ protocols and product labeling available publicly at Drugs@FDA. Drugs approved from 2016 to 2020 were analyzed. On the basis of publicly available study protocols and Food and Drug Administration (FDA) reviews, the authors reviewed the assessment of health related QoL in 53 clinical trials supporting 50 drug approvals from 2016 to 2020. Those trials resulted in approval of medications to treat leukemia, non-Hodgkin lymphoma, Hodgkin lymphoma, myelodysplastic syndrome, and multiple myeloma. Results: A total of 14,819 patients were assessed in the 5 years period after exclusion of unpublished studies. We calculated the frequency of assessment of health related QoL and if QoL measures were reported in the subsequent publications. After exclusion of 7 unpublished studies, we analyzed 46 clinical trials for reporting of HRQOL. Thirty percent of the protocols included assessment of health related QoL with only 43% of them (13% of all the analyzed clinical trials) reported the results of health related QoL. The results were consistent across all subtypes of hematological malignancies and were similar regardless of the studied primary endpoint. Conclusions: QoL measures are under studied in clinical trials of hematological malignancies. Studies that measured health related QoL didn’t report the results in more than half of the time. Although results may be reported in separate future publications, improvement in assessing and reporting of QoL will help to incorporate QoL measures in patient care and therapy choice decision. [Table: see text]
- Published
- 2020
36. HL-210: Assessment and Reporting of Quality of Life Measures in Pivotal Clinical Trials of Lymphoma
- Author
-
Carlos A. Ramos, Helen E. Heslop, Samer Al Hadidi, Rammurti T. Kamble, and George Carrum
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Follicular lymphoma ,Context (language use) ,Hematology ,medicine.disease ,humanities ,Clinical trial ,Oncology ,Quality of life ,Internal medicine ,Clinical endpoint ,Medicine ,T-cell lymphoma ,Mantle cell lymphoma ,business ,Diffuse large B-cell lymphoma - Abstract
Context Clinical trials are integral to improve treatment outcomes for patients with lymphoma. The importance of assessing and reporting of health-related quality of life (QoL) has been increasingly recognized in the field of oncology. QoL measures, which reflect patient's perceived benefits and satisfaction, might be especially important in lymphoma clinical trials, where the intervention may not result in cure. Objective To investigate assessment of health-related QoL in clinical trials that resulted in a subsequent approval of drugs to treat lymphoma by Food and Drug Administration (FDA) and to determine if ensuing published studies reported QoL as part of their results. Design Data on health-related QoL were obtained from studies' protocols and product labeling available publicly at Drugs@FDA. Drugs approved in the period of 2016 till 2020 were analyzed. Setting On the basis of publicly available study protocols and FDA reviews, the authors reviewed the assessment of health-related QoL in 19 clinical trials supporting 17 drug approvals. Those trials resulted in approval of medications to treat classic Hodgkin lymphoma, follicular lymphoma, marginal zone lymphoma, mantle cell lymphoma, diffuse large B cell lymphoma, and T cell lymphoma. Patients or other participants A total of 5292 patients were assessed in the 5-year period after exclusion of unpublished studies. Main outcome measures We calculated the frequency of assessment of health-related QoL and if QoL measures were reported in the subsequent publications. Results After exclusion of 2 unpublished studies, we analyzed 17 clinical trials for reporting of health-related QoL. Eighteen percent of the protocols included assessment of health-related QoL with only 33% of them (6% of all the analyzed clinical trials) reported the results of health-related QoL. The results were consistent across all lymphoma subtypes and were similar regardless of the studied primary endpoint. Conclusions Health-related QoL measures are under studied in clinical trials of lymphoma. Studies that measured QoL didn't report the results in more than half of the time. Improvement in assessing and reporting of QoL will help to incorporate QoL measures in patient care and therapy choice decision.
- Published
- 2020
37. Outcomes of myeloablative, T cell deplete unrelated donor hematopoietic stem cell transplantation at a single center
- Author
-
George Carrum, Hussein Hamad, Rammurti T. Kamble, Helen E. Heslop, Sai Ravi Pingali, Albert Jang, Mahmoud Gaballa, Carlos A. Ramos, Gloria Obi, Premal Lulla, Shaun Bulsara, Meng-Fen Wu, LaQuisa Hill, Audrey Scholoff, and Ngoc Vu
- Subjects
Cancer Research ,Hematopoietic cell ,business.industry ,Myeloablative conditioning ,T cell ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Single Center ,Transplantation ,medicine.anatomical_structure ,Oncology ,Unrelated Donor ,Cancer research ,medicine ,business - Abstract
e19525 Background: Myeloablative conditioning (MAC) and unrelated donor (UD) hematopoietic cell transplantation (HCT) are associated with increased non-relapse mortality (NRM) compared with matched related donor (MRD) HCT [18%, 21%, and 32% for MRD, Matched UD (MUD), and mismatched MUD (MMUD), respectively, p < 0.001] (Saber et al, 2012). Our center uses alemtuzumab as means of T cell depletion to mitigate NRM risk in UD HCT. Here, we report outcomes utilizing this strategy. Methods: We retrospectively analyzed adult patients (pts) who received alemtuzumab-based T cell deplete MAC UD HCT from August 2012 to December 2017. Outcomes were estimated via Kaplan Meier and cumulative incidence methods. Results: Sixty-eight pts underwent T cell deplete MAC UD HCT for AML/MDS (n = 32), ALL (n = 18), lymphoma (n = 5), and other diseases (n = 13). Thirty-nine (57.4%) underwent a MUD HCT, while 29 (42.4%) underwent a MMUD (≤ 9/10 match) HCT. Median follow up was 35.5 months (m) (range 1.5 – 71.1). Median age was 46 years (range 21 - 68), 58.8% were male. Nineteen AML pts (67.9% of all AML) were considered high risk defined as secondary AML (n = 3), CR2 or more (n = 9), primary induction failure (n = 6) or having gross residual disease at time of HCT (n = 1). At 24 m, overall survival (OS) for the entire cohort was 65.3% (95%CI (CI) 53.5%- 77.2%), GVHD relapse-free survival (GRFS) was 54.8% (CI 42.8%- 66.9%), NRM was 10.4% (CI 4.5%- 19.2%), and relapse incidence (RI) was 27.3% (CI 17.1%- 38.5%). None of the 68 pts had graft failure or rejection. In the AML/MDS cohort, at 24 m OS was 52.3% (CI 33.9%- 70.7%), GRFS was 41.4% (CI 23.9%- 59%), NRM was 9.7% (CI 2.4%- 23.2%), and RI was 39.1% (CI 21.8%- 56.2%). Non-AML/MDS pts at 24 m had OS, GRFS, NRM and RI of 76.8% (CI 62.6%- 90.9%), 66.7% (CI 51.3%- 82.08%), 11.1% (CI 3.4%- 23.9%), and 16.7% (CI 6.6%- 30.6%), respectively. The table summarizes our outcomes. Conclusions: We show that alemtuzumab-based MAC conditioning for UD HCT has the benefit of being well tolerated with low NRM and high GRFS, while having comparable RI and OS compared to published outcomes of T cell replete MAC UD HCT (Saber et al, 2012). However, the retrospective nature without a comparator is a major limitation which necessitates a future prospective study to validate these findings. [Table: see text]
- Published
- 2020
38. Efficacy of Afternoon Plerixafor Administration for Stem Cell Mobilization
- Author
-
Gloria Obi Anyadike, Rammurti T. Kamble, George Carrum, James E. Cox, Romelia May, Cynthia El Rahi, and Audrey Scholoff
- Subjects
Oncology ,medicine.medical_specialty ,Hematology ,Bone marrow transplantation ,Stem cell mobilization ,business.industry ,hematology ,lcsh:RC633-647.5 ,Plerixafor ,lcsh:Diseases of the blood and blood-forming organs ,03 medical and health sciences ,0302 clinical medicine ,Apheresis ,030220 oncology & carcinogenesis ,Internal medicine ,Peak effect ,oncology ,medicine ,business ,Hematopoietic Stem Cell Mobilization ,030215 immunology ,medicine.drug ,Original Research - Abstract
Background: When used for hematopoietic stem cell mobilization, plerixafor was originally recommended to be administered 11 hours prior to apheresis based on the peak effect of 10 to 14 hours translating into an administration time of 10 to 11 pm. Reports of post-plerixafor anaphylactic reactions mandated labeling change by the Food and Drug Administration with recommendation of monitoring patients after administration. Based on data suggesting sustained plerixafor activity at 18 hours, we changed our administration time to 4 pm at our center. Objective: The objective of this study is to compare the stem cell collection efficiency before and after the practice change at our institution. Methods: A retrospective chart review for patients with multiple myeloma, Hodgkin lymphoma, and non-Hodgkin lymphoma who received a plerixafor-containing mobilization regimen was conducted. The primary end point was the percentage of patients achieving the minimal CD34+ cell goal in ⩽2 apheresis days. The secondary end points included the percentage of patients achieving the preferred CD34+ cell goal in ⩽2 apheresis days, days of apheresis, total CD34+ cells Collected, and engraftment time. Results: A total of 208 patients (4 pm group n = 68, 10 pm group n = 140) with multiple myeloma (n = 112), Hodgkin lymphoma (n = 10), and non-Hodgkin lymphoma (n = 86) were included in the analysis. About 91% and 89% ( P = .804) of the patients in the 4 and 10 pm groups, respectively, collected minimum cell dose. Preferred CD34+ cell goal was achieved in 57% and 53% of patients in the 4 and 10 pm groups, respectively. Conclusions: Late afternoon administration of plerixafor provides efficient stem cell mobilization.
- Published
- 2018
39. Voriconazole photosensitivity causes apparent graft-versus-host disease exacerbation
- Author
-
Rammurti T. Kamble, Ngoc Vu, Gloria Obi, Akash Mukherjee, Audrey Scholoff, Ekim Ekinci, and George Carrum
- Subjects
Adult ,Male ,medicine.medical_specialty ,Antifungal Agents ,Exacerbation ,MEDLINE ,Graft vs Host Disease ,03 medical and health sciences ,0302 clinical medicine ,Photosensitivity ,medicine ,Humans ,Photosensitivity Disorders ,Aged ,Voriconazole ,Transplantation ,business.industry ,Hematology ,Middle Aged ,medicine.disease ,Dermatology ,Graft-versus-host disease ,030220 oncology & carcinogenesis ,Female ,business ,030215 immunology ,medicine.drug - Published
- 2018
40. Olanzapine for Chemotherapy-Induced Nausea and Vomiting (CINV) Prophylaxis in Patients Receiving High-Dose Chemotherapy for Autologous Hematopoietic Stem Cell Transplantation (ASCT)
- Author
-
Jade L. Hefler, Premal Lulla, George Carrum, Carlos A. Ramos, Rammurti T. Kamble, James E. Cox, Joe Ensor, Edward T. McLean, and Ekim Ekinci
- Subjects
Melphalan ,Olanzapine ,Transplantation ,medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Hematology ,Hematopoietic stem cell transplantation ,humanities ,Fosaprepitant ,Ondansetron ,Regimen ,Internal medicine ,medicine ,business ,medicine.drug ,Chemotherapy-induced nausea and vomiting - Abstract
Introduction Olanzapine has demonstrated clinical efficacy for CINV prophylaxis. Efficacy and safety of olanzapine for CINV prophylaxis in patients receiving high-dose chemotherapy as conditioning regimen for HSCT is not established. Objective The primary objective is to compare efficacy and safety of olanzapine versus fosaprepitant for CINV prophylaxis in patients receiving high-dose chemotherapy for ASCT. Methods A single-center, retrospective analysis evaluated consecutive patients who received high-dose melphalan or BEAM for ASCT from June 2016 to September 2018. April 2017 onwards, we switched CINV prophylaxis to olanzapine, replacing fosaprepitant. In this analysis, outcomes of an olanzapine regimen combined with ondansetron and dexamethasone (OOD) were compared with fosaprepitant combined with ondansetron and dexamethasone (FOD). Assessment period for CINV was defined as acute phase (chemotherapy days) and delayed phase (within 5 days of chemotherapy completion). The primary endpoint is the proportion of days requiring rescue CINV medications during the assessment period. Secondary endpoints include number of rescue medication doses required during the acute and delayed phases, time to neutrophil engraftment (TTNE), and length of stay (LOS). A cost-effectiveness analysis is being performed. Results A total of 154 patients were evaluated (FOD = 83 and OOD = 71). Patient demographics were similar in both groups, except that patients in the OOD cohort were older (median age 63 vs. 55 years; p = 0.0002). Patients received the following conditioning regimens: melphalan 140 mg/m2 (FOD = 10; OOD = 21), melphalan 200 mg/m2 (FOD = 28; OOD = 44), BEAM (FOD = 45; OOD = 6). In the acute phase, 10% of FOD patients required rescue CINV medications on at least 50% of days while approximately 3% of OOD patients required rescue medications on any day (p = 0.006). In the delayed phase, the median proportion of days requiring rescue CINV medications was significantly lower with OOD (60% vs. 20%; p Conclusion Olanzapine-based CINV prophylaxis was significantly superior to fosaprepitant for the proportion of days requiring rescue CINV medications.
- Published
- 2019
41. Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity
- Author
-
Yasmin Hazrat, Carlos A. Ramos, Meng Fen Wu, Aaron E. Foster, Barbara Savoldo, Paul Castillo-Caro, A. John Barrett, Malcolm K. Brenner, Alana A. Kennedy-Nasser, Hao Liu, George Carrum, Catherine M. Bollard, Robert A. Krance, Jos Melenhorst, Stephanie Ku, Kathryn S. Leung, Helen E. Heslop, Eric Yvon, and Sawa Ito
- Subjects
Male ,Cancer Research ,Adolescent ,medicine.medical_treatment ,Population ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Hematopoietic stem cell transplantation ,Biology ,Antiviral Agents ,T-Lymphocytes, Regulatory ,Article ,Antigen ,medicine ,Humans ,Transplantation, Homologous ,Lymphocyte Count ,Prospective Studies ,IL-2 receptor ,Child ,education ,Cell Proliferation ,education.field_of_study ,Leukemia ,Dose-Response Relationship, Drug ,Hematopoietic Stem Cell Transplantation ,Cancer ,FOXP3 ,Interleukin ,Flow Cytometry ,medicine.disease ,Treatment Outcome ,Oncology ,Immunology ,Interleukin-2 ,Female - Abstract
Purpose: GVHD after allogeneic hematopoietic stem cell transplantation (alloSCT) has been associated with low numbers of circulating CD4+CD25+FoxP3+ regulatory T cells (Tregs). Because Tregs express high levels of the interleukin (IL)-2 receptor, they may selectively expand in vivo in response to doses of IL-2 insufficient to stimulate T effector T-cell populations, thereby preventing GVHD. Experimental Design: We prospectively evaluated the effects of ultra low-dose (ULD) IL-2 injections on Treg recovery in pediatric patients after alloSCT and compared this recovery with Treg reconstitution post alloSCT in patients without IL-2. Sixteen recipients of related (n = 12) or unrelated (n = 4) donor grafts received ULD IL-2 post hematopoietic stem cell transplantation (HSCT; 100,000–200,000 IU/m2 ×3 per week), starting Results: No grade 3/4 toxicities were associated with ULD IL-2. CD4+CD25+FoxP3+ Tregs increased from a mean of 4.8% (range, 0%–11.0%) pre IL-2 to 11.1% (range, 1.2%–31.1%) following therapy, with the greatest change occurring in the recipients of matched related donor (MRD) transplants. No IL-2 patients developed grade 2–4 acute GVHD (aGVHD), compared with 4 of 33 (12%) of the comparator group who did not receive IL-2. IL-2 recipients retained T cells reactive to viral and leukemia antigens, and in the MRD recipients, only 2 of 13 (15%) of the IL-2 patients developed viral infections versus 63% of the comparator group (P = 0.022). Conclusions: Hence, ULD IL-2 is well tolerated, expands a Treg population in vivo, and may be associated with a lower incidence of viral infections and GVHD. Clin Cancer Res; 20(8); 2215–25. ©2014 AACR.
- Published
- 2014
42. Sustained Complete Responses in Patients With Lymphoma Receiving Autologous Cytotoxic T Lymphocytes Targeting Epstein-Barr Virus Latent Membrane Proteins
- Author
-
Andrea M. Sheehan, Youli Zu, Elizabeth J. Shpall, Cliona M. Rooney, Barbara Pro, Stephanie Ku, Daniel Lee, George Carrum, Oumar Diouf, Malcolm K. Brenner, Meng Fen Wu, Hao Liu, Catherine M. Bollard, Adrian P. Gee, Luis Fayad, Stephen Gottschalk, Yasmin Hazrat, Carlos A. Ramos, Helen E. Heslop, and Vicky Torrano
- Subjects
Adult ,Male ,Herpesvirus 4, Human ,Cancer Research ,Time Factors ,Adolescent ,Lymphoma ,medicine.medical_treatment ,Genetic Vectors ,Kaplan-Meier Estimate ,medicine.disease_cause ,Immunotherapy, Adoptive ,Transplantation, Autologous ,Disease-Free Survival ,Virus ,Adenoviridae ,Cell Line ,Viral vector ,Viral Matrix Proteins ,Young Adult ,Antigen ,Recurrence ,Risk Factors ,Transduction, Genetic ,medicine ,Humans ,Cytotoxic T cell ,Child ,Aged ,Cell Proliferation ,business.industry ,Remission Induction ,Genetic Therapy ,Immunotherapy ,Middle Aged ,medicine.disease ,Texas ,Epstein–Barr virus ,Transplantation ,Treatment Outcome ,Oncology ,Immunology ,Female ,business ,T-Lymphocytes, Cytotoxic - Abstract
Purpose Tumor cells from approximately 40% of patients with Hodgkin or non-Hodgkin lymphoma express the type II latency Epstein-Barr virus (EBV) antigens latent membrane protein 1 (LMP1) and LMP2, which represent attractive targets for immunotherapy. Because T cells specific for these antigens are present with low frequency and may be rendered anergic by the tumors that express them, we expanded LMP–cytotoxic T lymphocytes (CTLs) from patients with lymphoma using autologous dendritic cells and EBV-transformed B–lymphoblastoid cell lines transduced with an adenoviral vector expressing either LMP2 alone (n = 17) or both LMP2 and ΔLMP1 (n = 33). Patients and Methods These genetically modified antigen-presenting cells expanded CTLs that were enriched for specificity against type II latency LMP antigens. When infused into 50 patients with EBV-associated lymphoma, the expanded CTLs did not produce infusional toxicities. Results Twenty-eight of 29 high-risk or multiple-relapse patients receiving LMP-CTLs as adjuvant therapy remained in remission at a median of 3.1 years after CTL infusion. None subsequently died as a result of lymphoma, but nine succumbed to complications associated with extensive prior chemoradiotherapy, including myocardial infarction and secondary malignancies. Of 21 patients with relapsed or resistant disease at the time of CTL infusion, 13 had clinical responses, including 11 complete responses. T cells specific for LMP as well as nonviral tumor-associated antigens (epitope spreading) could be detected in the peripheral blood within 2 months after CTL infusion, but this evidence for epitope spreading was seen only in patients achieving clinical responses. Conclusion Autologous T cells directed to the LMP2 or LMP1 and LMP2 antigens can induce durable complete responses without significant toxicity. Their earlier use in the disease course may reduce delayed treatment-related mortality.
- Published
- 2014
43. Effect of Routine Surveillance Imaging on the Outcomes of Patients With Classical Hodgkin Lymphoma After Autologous Hematopoietic Cell Transplantation
- Author
-
Kristin Richardson, Namrata Shah, Timothy S. Fenske, Parameswaran Hari, Sai Ravi Pingali, Narendranath Epperla, Mehdi Hamadani, Reem Karmali, George Carrum, and Jonathan Kapke
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,Transplantation, Autologous ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Refractory ,medicine ,Classical Hodgkin lymphoma ,Humans ,In patient ,Aged ,Retrospective Studies ,Hematopoietic cell ,business.industry ,Hematopoietic Stem Cell Transplantation ,Retrospective cohort study ,Hematology ,Middle Aged ,medicine.disease ,Hodgkin Disease ,Surgery ,Lymphoma ,Transplantation ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Female ,Radiology ,Surveillance imaging ,business ,030215 immunology - Abstract
Background Patients with relapsed and refractory classical Hodgkin lymphoma (cHL) are often treated with autologous hematopoietic cell transplantation (auto-HCT). After auto-HCT, most transplant centers implement routine surveillance imaging to monitor for disease relapse; however, there is limited evidence to support this practice. Patients and Methods In this multicenter, retrospective study, we identified cHL patients (n = 128) who received auto-HCT, achieved complete remission (CR) after transplantation, and then were followed with routine surveillance imaging. Of these, 29 (23%) relapsed after day 100 after auto-HCT. Relapse was detected clinically in 14 patients and with routine surveillance imaging in 15 patients. Results When clinically detected relapse was compared with to radiographically detected relapse respectively, the median overall survival (2084 days [range, 225-4161] vs. 2737 days [range, 172-2750]; P = .51), the median time to relapse (247 days [range, 141-3974] vs. 814 days [range, 96-1682]; P = .30) and the median postrelapse survival (674 days [range, 13-1883] vs. 1146 days [range, 4-2548]; P = .52) were not statistically different. In patients who never relapsed after auto-HCT, a median of 4 (range, 1-25) surveillance imaging studies were performed over a median follow-up period of 3.5 years. Conclusion A minority of patients with cHL who achieve CR after auto-HCT will ultimately relapse. Surveillance imaging detected approximately half of relapses; however, outcomes were similar for those whose relapse was detected using routine surveillance imaging versus detected clinically in between surveillance imaging studies. There appears to be limited utility for routine surveillance imaging in cHL patients who achieve CR after auto-HCT.
- Published
- 2016
44. T Cell Immunity Toward Viral- and Tumor-Antigens Is Preserved in MDS Patients
- Author
-
Rammurti T. Kamble, Premal Lulla, Spyridoula Vasileiou, Quillan Huang, George Carrum, Hussein Hamad, LaQuisa Hill, Shivani Mukhi, Ann M. Leen, Wingchi K Leung, and Carlos A. Ramos
- Subjects
business.industry ,medicine.medical_treatment ,Immunology ,Cytomegalovirus ,Cell Biology ,Hematology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Pancytopenia ,Cytokine ,Antigen ,Immunity ,Survivin ,medicine ,Interleukin 8 ,business ,Cyclin - Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of disorders characterized by bone marrow failure and a propensity to progress to acute myeloid leukemia (AML). A core component of the underlying pathogenesis in MDS is deregulation of inflammatory cytokines, such as tumor growth factor-β (TGFβ), which impact the function of immune cells and hence their capacity to mount anti-infective or anti-tumor responses. However, little is known about antigen-specific T cell function in patients with MDS. We hypothesized that virus-specific T cell (VST) function might be preserved in patients with MDS, and that the functional capacity of T cells reactive against tumor-associated antigens aberrantly overexpressed by clonal MDS cells such as Cyclin A1 (CCNA1) and Proteinase (PR3) might also be preserved and exploited for immunotherapeutic purposes. Following informed consent, we collected peripheral blood samples from 10 patients (pts) with MDS and 17 healthy donors. Most pts (9 out of 10) were transfusion dependent and 3 subsequently underwent an allogeneic HSCT. Table 1 summarizes the other clinical characteristics, karyotypic and mutational profile at the time of blood collection. Compared with T cells isolated from healthy donors, MDS patient-derived T cells had a similar CD4 to CD8 ratio (1.5-2.5:1 for healthy donors and 3:1 for MDS pts), but displayed a more exhausted profile at baseline (CD3+TIM3+: 1% in healthy donors and 5% in MDS pts) and produced higher levels of inflammatory cytokines [IFNγ (18±3pg/ml vs 36±16pg/ml, healthy donor vs MDS; p=0.12), and IL-8 (56±32 vs 704±446 pg/ml, p=0.01)]. Next, to assess the capacity of MDS pts to mount ex vivo functional virus-directed responses, we stimulated patient-derived PBMCs (n=5) with overlapping peptide libraries (pepmixes) spanning immunogenic AdV, CMV, EBV, BK and HHV-6 antigens. Similar to healthy donor-derived T cell lines (n=5, 3 specific for 4 viruses and 2 for 5 viruses), all 5 MDS patient-derived lines demonstrated specificity for one or more of the target viruses (1 for 5 viruses, 1 for 4, 2 for 3 and 1 for 1 virus) as observed by IFNγ ELISpot assay with comparable magnitude (range Adv: 43-730 vs 384-941 in healthy donors, CMV: 0-1599 vs 0-3002, EBV: 0-1486 vs 0-541, BK: 0-839 vs 38-275 and HHV6: 0-794 vs 5-407 SFU/2x105 cells, respectively). We next examined the feasibility of expanding autologous MDS-antigen directed T cell products (n=10) to determine whether an adoptive immunotherapeutic approach might be applicable for MDS treatment. Thus, we exposed patient-derived PBMCs to autologous dendritic cells (DC) loaded with pepmixes spanning 6 MDS-associated antigens (CCNA1, survivin, WT1, PRAME, PR3 and NYESO1). After 3 rounds of stimulation, the products obtained were predominantly CD3+ T cells (mean 88±1.3%) that were polyclonal (CD4: 46±5% and CD8: 41±4%) containing predominantly memory T cells (TEM: 36±6% TCM: 37±5% and Tnaïve =13±3%). Six lines (60%) showed specific recognition to at least one of the target antigens: 4 lines specific for PRAME, 1 for CCNA1, 1 for WT1 and 1 for NYESO1 (range 0-40, 0-184, 0-1386 and 0-179 SFU/2x105 cells, respectively by IFNγ ELIspot). T cell lines were capable of specifically secreting multiple effector cytokines in response to targets (TNFα: 12% and IFNγ: 16% in response to PRAME in a representative patient-derived T cell line). Furthermore, this line was capable of killing PRAME+ targets in a 4hr 51Cr release assay [60% specific lysis, E:T 20:1]. In conclusion, functional virus-directed T cell immunity in patients with MDS is preserved, potentially explaining the lower rates of viral reactivation seen in these patients compared with other infections. Moreover, T cells specific for MDS-expressed tumor antigens can also be successfully expanded ex vivo from patients. Taken together this raises the possibility of applying an adoptive immunotherapeutic approach for the treatment of MDS. Disclosures Ramos: Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Tessa Therapeutics: Research Funding. Leen:Allovir: Consultancy, Other: Cofounder, Ownership Interest; Marker Therapeutics: Consultancy, Other: Cofounder, Ownership Interest.
- Published
- 2019
45. The Impact of Donor Baseline Characteristics on Total Nucleated Cell Count in Marrow Products of Healthy Bone Marrow Donors
- Author
-
Brennan M. Parmelee, Premal Lulla, Josie Sena, LaQuisa Hill, Gloria Obi Anyadike, George Carrum, Audrey Scholoff, Kimberly East, Carlos A. Ramos, Helen E. Heslop, Marsha Cohen, and Jaya Paranilam
- Subjects
Oncology ,Transplantation ,Univariate analysis ,medicine.medical_specialty ,business.industry ,Bone marrow donors ,Hematopoietic stem cell ,Hematology ,Single Center ,medicine.anatomical_structure ,Baseline characteristics ,Internal medicine ,medicine ,business - Abstract
Introduction An adequate dose of marrow derived total nucleated cells (TNCs) is essential for engraftment in recipients undergoing allogeneic hematopoietic stem cell (HSC) transplant. Marrow collection centers anticipate the harvest volume required to meet a minimum collection goal of 2 × 108 TNCs/kg based on published aggregates of donor marrow TNC concentrations (currently set at 18.3 × 109/L). However, marrow TNCs are highly variable among marrow donors. We hypothesized that baseline donor characteristics such as age, BMI, and ethnicity, could be used to more accurately predict TNCs. Objectives We commissioned an IRB-approved retrospective single center study to ascertain which donor characteristics could independently predict marrow TNCs during a harvest as well as the impact on successful recipient engraftment. Methods Univariate linear regression was conducted to estimate the crude association between each factor and TNC (487 donors). The following variables were considered as possible predictors: donor BMI, age, ethnicity, gender, and total harvest volume. Variables that had a p value Results Univariate analysis revealed no difference in TNCs between genders (p = 0.697). BMI was significantly positively correlated with TNCs (t = 4.17; p Conclusion Our findings demonstrate that multiple donor characteristics can predict marrow TNCs. The impact of ethnicity on marrow TNC counts, but not on engraftment rates, suggests that the true HSC concentrations might be higher in these donors despite the lower TNCs. As donor pools diversify and use of haploidentical donors increases, our observations indicate that expected TNC counts during marrow harvests will vary based on baseline donor characteristics.
- Published
- 2019
46. Targeting Lymphomas Using Non-Engineered, Multi-Antigen-Specific T Cells
- Author
-
Manik Kuvalekar, Mrinalini Bilgi, Ifigeneia Tzannou, Rayne H. Rouce, Adrian P. Gee, Cliona M. Rooney, Bambi Grilley, Carlos A. Ramos, Malcolm K. Brenner, Ann M. Leen, George Carrum, Juan F. Vera, Rammurti T. Kamble, Tao Wang, Ayumi Watanabe, Premal Lulla, and Helen E. Heslop
- Subjects
Transplantation ,Adoptive cell transfer ,PRAME ,biology ,business.industry ,medicine.medical_treatment ,T cell ,Hematology ,Immunotherapy ,CD19 ,medicine.anatomical_structure ,Antigen ,medicine ,biology.protein ,Cancer research ,business ,B cell ,CD8 - Abstract
Immunotherapy is emerging as a potent treatment for a range of hematologic malignancies including lymphomas. Indeed adoptive transfer of T cells genetically engineered to express the CD19 CAR has now received FDA approval for the treatment of refractory diffuse large B cell lymphomas (DLBCL). We have developed a non-engineered T cell therapy to treat patients with Hodgkin's and non-Hodgkin's lymphoma (HL/NHL) using single T cell lines that simultaneously target the tumor-associated antigens (TAAs) PRAME, SSX2, MAGEA4, NY-ESO-1 and Survivin. We can consistently prepare these lines by culturing PBMCs in the presence of a Th1-polarizing/pro-proliferative cytokine cocktail, and adding autologous pepmix-loaded DCs as APCs. The use of whole antigen overcomes the HLA restriction imposed by the use of transgenic TCRs specific for single peptides, while targeting multiple antigens simultaneously should reduce the risk of tumor immune evasion. We have generated 42 clinical-grade multiTAA T cell lines, comprising CD3+ T cells (mean 98±1.1%) with a mixture of CD4+ (mean 48±4.3%) and CD8+ (mean 37±4%) T cells that recognize the targeted antigens PRAME, SSX2, MAGEA4, NY-ESO-1 and Survivin (range 0-463, 0-496, 0-330, 0-379 and 0-304 SFU/2 × 105, respectively in IFNg ELIspot). None of the lines reacted against non-malignant autologous cells (3±3.8% specific lysis; E:T 20:1). We have treated 33 patients: 13 with HL, 17 with aggressive NHL (DLBCL, mantle cell, T cell lymphomas) and 3 with indolent NHLs (FL, marginal zone lymphoma). Patients were infused with 0.5-2 × 107 multiTAA-T cells/m2 and did not receive lymphodepleting chemotherapy. Of 18 patients who were infused as adjuvant all but 2 remain in remission (range 3-42 months). Fifteen patients have received multiTAA-specific T cells to treat active disease, all of whom had failed a median of 4 lines of prior therapy. Of these, 5 had transient disease stabilization followed by disease progression, 4 have ongoing stable disease at 3-18 months post-multiTAA-specific T cells while the remaining 6 (3 with HL and 3 with DLBCL) had complete and durable responses (4 to 41 months), as assessed by PET. These clinical responses correlated with the detection of tumor-reactive T cells in patient peripheral blood post-infusion directed against both targeted antigens as well as non-targeted TAAs including MAGEA2B and MAGE C1, indicating induction of antigen/epitope spreading. Notably, no patient, including the complete responders, had infusion-related systemic- or neuro-toxicity. Thus, infusion of autologous multiTAA-targeted T cells directed to PRAME, SSX2, MAGEA4, NY-ESO-1 and Survivin has been safe, and despite concerns that endogenous T cells directed against shared/cancer testis antigens would be of insufficient affinity to produce prolonged benefit our multiTAA-specific T cells were able to induce durable complete responses in patients with lymphomas.
- Published
- 2019
47. Efficacy of deferred dosing of granulocyte colony-stimulating factor in autologous hematopoietic transplantation for multiple myeloma
- Author
-
Rammurti T. Kamble, George Carrum, Malcolm K. Brenner, Hao Liu, R. May, Helen E. Heslop, S. Campos, Jesse Wu, J. E. Cox, and Carlos A. Ramos
- Subjects
autologous transplant ,Adult ,Male ,medicine.medical_specialty ,Cost effectiveness ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,G-CSF ,Transplantation, Autologous ,Granulocyte Colony-Stimulating Factor ,medicine ,Mucositis ,Humans ,Dosing ,Aged ,Retrospective Studies ,Transplantation ,Neutrophil Engraftment ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,Middle Aged ,medicine.disease ,Granulocyte colony-stimulating factor ,Surgery ,multiple myeloma ,Treatment Outcome ,Anesthesia ,Original Article ,Female ,business ,Febrile neutropenia - Abstract
Routine administration of G-CSF following autologous hematopoietic SCT (ASCT) expedites ANC recovery and reduces hospitalization by 1–2 days; it has no impact on febrile neutropenia, infections, morbidity, mortality, event-free survival or OS. To determine whether delayed G-CSF dosage could result in equivalent ANC recovery and thereby improve cost effectiveness, we deferred the administration of G-CSF until WBC recovery had begun. A total of 117 patients with multiple myeloma received ASCT from January 2005 to September 2012. Of these, 52 were in the conventional dosing group (CGD) and received G-CSF from Day +7 for a median of five doses. In the deferred dosing group (DGD), 65 patients received G-CSF from median day 14 post transplant for a median of zero doses. There was no difference between groups in the incidence or duration of febrile neutropenia, duration of ⩾grade III mucositis, weight gain, rash, engraftment syndrome or early death (100 days). The DGD group had a significantly longer time to neutrophil engraftment than the CGD group (15 days vs 12 days; P
- Published
- 2013
48. Donor body mass index is an important factor that affects peripheral blood progenitor cell yield in healthy donors after mobilization with granulocyte-colony-stimulating factor
- Author
-
Martha Kennedy, Sandra Potts, George Carrum, Christopher Leveque, Salvador Garcia, Kevin M. Burns, Jian Chen, Francisco J. Segura, and Aleksandar Babic
- Subjects
Mobilization ,business.industry ,medicine.medical_treatment ,Immunology ,CD34 ,Hematology ,Hematopoietic stem cell transplantation ,Leukapheresis ,Granulocyte colony-stimulating factor ,Transplantation ,Andrology ,Cord blood ,medicine ,Immunology and Allergy ,Progenitor cell ,business - Abstract
Background The use of hematopoietic progenitor cell (HPC) transplantation has rapidly expanded in recent years. Currently, several sources of HPCs are available for transplantation including peripheral blood HPCs (PBPCs), cord blood cells, and marrow cells. Of these, PBPC collection has become the major source of HPCs. An important variable in PBPC collection is the response to PBPC mobilization, which varies significantly and sometime causes mobilization failure. Study Design and Methods A retrospective study of 69 healthy donors who underwent PBPC donation by leukapheresis was performed. All of these donors received 10 μg/kg/day or more granulocyte–colony-stimulating factor (G-CSF) for 5 days before PBPC harvest. Donor factors were evaluated and correlated with mobilization responses, as indicated by the precollection CD34 count (pre-CD34). Results Donors with a pre-CD34 of more than 100 × 106/L had higher body mass index (BMI) compared with donors whose pre-CD34 was 38 × 106 to 99 × 106/L or less than 38 × 106/L (32.0 ± 1.04 kg/m2 vs. 28.7 ± 0.93 kg/m2 vs. 25.9 ± 1.27 kg/m2, respectively; p
- Published
- 2013
49. Outcomes and costs of autologous stem cell mobilization with chemotherapy plus G-CSF vs G-CSF alone
- Author
-
Guido Tricot, Stephen Brown, Nelson J. Chao, Leona Holmberg, Junya Kanda, Lisa M. Bernard, Paul J. Shaughnessy, Jane L. Liesveld, Mitchell E. Horwitz, Anthony D. Sung, Daniel T. Grima, Brian McClune, and George Carrum
- Subjects
Male ,Lymphoma ,medicine.medical_treatment ,Mobilization ,Hematopoietic stem cell transplantation ,Gastroenterology ,Chemo-mobilization ,Antibodies, Monoclonal, Murine-Derived ,0302 clinical medicine ,Multiple myeloma ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Granulocyte Colony-Stimulating Factor ,Etoposide ,Hematopoietic Stem Cell Transplantation ,Hematology ,Middle Aged ,Hematopoietic Stem Cell Mobilization ,3. Good health ,Granulocyte colony-stimulating factor ,Treatment Outcome ,030220 oncology & carcinogenesis ,Female ,Rituximab ,medicine.drug ,Adult ,medicine.medical_specialty ,Cyclophosphamide ,G-CSF ,Transplantation, Autologous ,Article ,Young Adult ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Aged ,Retrospective Studies ,Transplantation ,Chemotherapy ,business.industry ,medicine.disease ,Surgery ,Autologous hematopoietic stem cell transplantation ,business ,Febrile neutropenia ,030215 immunology - Abstract
Chemotherapy plus G-CSF (C+G) and G-CSF alone are two of the most common methods used to mobilize CD34(+) cells for autologous hematopoietic SCT (AHSCT). In order to compare and determine the real-world outcomes and costs of these strategies, we performed a retrospective study of 226 consecutive patients at 11 medical centers (64 lymphoma, 162 multiple myeloma), of whom 55% of lymphoma patients and 66% of myeloma patients received C+G. Patients with C+G yielded more CD34(+) cells/day than those with G-CSF alone (lymphoma: average 5.51 × 10(6) cells/kg on day 1 vs 2.92 × 10(6) cells/kg, P=0.0231; myeloma: 4.16 × 10(6) vs 3.69 × 10(6) cells/kg, P0.00001) and required fewer days of apheresis (lymphoma: average 2.11 vs 2.96 days, P=0.012; myeloma: 2.02 vs 2.83 days, P=0.0015), although nearly all patients ultimately reached the goal of 2 × 10(6) cells/kg. With the exception of higher rates of febrile neutropenia in myeloma patients with C+G (17% vs 2%, P0.05), toxicities and other outcomes were similar. Mobilization with C+G cost significantly more (lymphoma: median $10,300 vs $7300, P0.0001; myeloma: $8800 vs $5600, P0.0001), although re-mobilization adds $6700 for drugs alone. Our results suggest that although both C+G and G-CSF alone are effective mobilization strategies, C+G may be more cost-effective for patients at high risk of insufficient mobilization.
- Published
- 2013
50. T Cell Therapy for Multiple Myeloma
- Author
-
Ann M. Leen, Rammurti T. Kamble, Ulrike Gerdemann, Hao Liu, Juan F. Vera, Bambi Grilley, Malcolm K. Brenner, Premal Lulla, Helen E. Heslop, Adrian P. Gee, and George Carrum
- Subjects
Transplantation ,medicine.anatomical_structure ,business.industry ,Myeloma protein ,T cell ,medicine ,Cancer research ,Hematology ,medicine.disease ,business ,Multiple myeloma - Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.