111 results on '"Stephen Gottschalk"'
Search Results
2. Paediatric Strategy Forum for medicinal product development of chimeric antigen receptor T-cells in children and adolescents with cancer
- Author
-
Elizabeth Fox, Michael Berntgen, Alfonso Quintás-Cardama, Veronique Minard-Colin, Susan L. Weiner, André Baruchel, Laura Pearce, Karsten Nysom, Andrew D.J. Pearson, Danielle H. Taylor, Christian M. Zwaan, Nirali N. Shah, Yousif Matloub, Ivan D. Horak, Gregory H. Reaman, Gilles Vassal, Koen Norga, Claudia Rossig, Dominik Karres, Lori A. Ehrlich, Martina Schüßler-Lenz, Alberto S. Pappo, Joe McDonough, Martina A. Sersch, Nick Richardson, Donna Ludwinski, Abraham Bassan, Eric Bleickardt, Nicole Scobie, Stephen Gottschalk, Rob Pieters, Sarah K. Tasian, Courtney Johnson, Teresa de Rojas, Malcolm A. Smith, Franca Ligas, Lynley V. Marshall, Shannon L. Maude, Brenda J. Weigel, Nick Bird, Najat Bouchkouj, Behzad K. Masouleh, Sarah Beaussant Cohen, Delphine Heenen, Rosanna Ricafort, G. Lesa, Linda Hanssens, Peter F. Bross, Carrie Brownstein, Crystal L. Mackall, Martin Pule, Douglas S. Hawkins, Jaroslav Sterba, and Maksim Mamonkin
- Subjects
0303 health sciences ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,medicine.medical_treatment ,Population ,Immunotherapy ,Disease ,Chimeric antigen receptor ,3. Good health ,Transplantation ,Cell therapy ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Drug development ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,Intensive care medicine ,education ,030304 developmental biology - Abstract
The seventh multi-stakeholder Paediatric Strategy Forum focused on chimeric antigen receptor (CAR) T-cells for children and adolescents with cancer. The development of CAR T-cells for patients with haematological malignancies, especially B-cell precursor acute lymphoblastic leukaemia (BCP-ALL), has been spectacular. However, currently, there are scientific, clinical and logistical challenges for use of CAR T-cells in BCP-ALL and other paediatric malignancies, particularly in acute myeloid leukaemia (AML), lymphomas and solid tumours. The aims of the Forum were to summarise the current landscape of CAR T-cell therapy development in paediatrics, too identify current challenges and future directions, with consideration of other immune effector modalities and ascertain the best strategies to accelerate their development and availability to children. Although the effect is of limited duration in about half of the patients, anti-CD19 CAR T-cells produce high response rates in relapsed/refractory BCP-ALL and this has highlighted previously unknown mechanisms of relapse. CAR T-cell treatment as first- or second-line therapy could also potentially benefit patients whose disease has high-risk features associated with relapse and failure of conventional therapies. Identifying patients with very early and early relapse in whom CAR T-cell therapy may replace haematopoietic stem cell transplantation and be definitive therapy versus those in whom it provides a more effective bridge to haematopoietic stem cell transplantation is a very high priority. Development of approaches to improve persistence, either by improving T cell fitness or using more humanised/fully humanised products and co-targeting of multiple antigens to prevent antigen escape, could potentially further optimise therapy. Many differences exist between paediatric B-cell non-Hodgkin lymphomas (B-NHL) and BCP-ALL. In view of the very small patient numbers with relapsed lymphoma, careful prioritisation is needed to evaluate CAR T-cells in children with Burkitt lymphoma, primary mediastinal B cell lymphoma and other NHL subtypes. Combination trials of alternative targets to CD19 (CD20 or CD22) should also be explored as a priority to improve efficacy in this population. Development of CD30 CAR T-cell immunotherapy strategies in patients with relapsed/refractory Hodgkin lymphoma will likely be most efficiently accomplished by joint paediatric and adult trials. CAR T-cell approaches are early in development for AML and T-ALL, given the unique challenges of successful immunotherapy actualisation in these diseases. At this time, CD33 and CD123 appear to be the most universal targets in AML and CD7 in T-ALL. The results of ongoing or planned first-in-human studies are required to facilitate further understanding. There are promising early results in solid tumours, particularly with GD2 targeting cell therapies in neuroblastoma and central nervous system gliomas that represent significant unmet clinical needs. Further understanding of biology is critical to success. The comparative benefits of autologous versus allogeneic CAR T-cells, T-cells engineered with T cell receptors T-cells engineered with T cell receptor fusion constructs, CAR Natural Killer (NK)-cell products, bispecific T-cell engager antibodies and antibody-drug conjugates require evaluation in paediatric malignancies. Early and proactive academia and multi-company engagement are mandatory to advance cellular immunotherapies in paediatric oncology. Regulatory advice should be sought very early in the design and preparation of clinical trials of innovative medicines, for which regulatory approval may ultimately be sought. Aligning strategic, scientific, regulatory, health technology and funding requirements from the inception of a clinical trial is especially important as these are very expensive therapies. The model for drug development for cell therapy in paediatric oncology could also involve a ‘later stage handoff’ to industry after early development in academic hands. Finally, and very importantly, strategies must evolve to ensure appropriate ease of access for children who need and could potentially benefit from these therapies.
- Published
- 2022
3. Transient blockade of TBK1/IKKε allows efficient transduction of primary human natural killer cells with vesicular stomatitis virus G-pseudotyped lentiviral vectors
- Author
-
Peter Chockley, Stephen Gottschalk, and Sagar L. Patil
- Subjects
0301 basic medicine ,Yellow fluorescent protein ,Cancer Research ,Cell signaling ,medicine.medical_treatment ,Genetic Vectors ,Immunology ,Protein Serine-Threonine Kinases ,Article ,Flow cytometry ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Transduction, Genetic ,medicine ,Animals ,Humans ,Immunology and Allergy ,Genetics (clinical) ,Transplantation ,biology ,medicine.diagnostic_test ,Chemistry ,Lentivirus ,Cell Biology ,Immunotherapy ,biology.organism_classification ,In vitro ,I-kappa B Kinase ,Genetically modified organism ,Cell biology ,Killer Cells, Natural ,030104 developmental biology ,Oncology ,Vesicular stomatitis virus ,030220 oncology & carcinogenesis ,Leukocytes, Mononuclear ,biology.protein ,Vesicular Stomatitis - Abstract
Background aims Vesicular stomatitis virus G (VSV-G)-pseudotyped lentiviral vectors (LVs) are widely used to reliably generate genetically modified, clinical-grade T-cell products. However, the results of genetically modifying natural killer (NK) cells with VSV-G LVs have been variable. The authors explored whether inhibition of the IKK-related protein kinases TBK1 and IKKe, key signaling molecules of the endosomal TLR4 pathway, which is activated by VSV-G, would enable the reliable transduction of NK cells by VSV-G LVs. Methods The authors activated NK cells from peripheral blood mononuclear cells using standard procedures and transduced them with VSV-G LVs encoding a marker gene (yellow fluorescent protein [YFP]) or functional genes (chimeric antigen receptors [CARs], co-stimulatory molecules) in the presence of three TBK1/IKKe inhibitors (MRT67307, BX-795, amlexanox). NK cell transduction was evaluated by flow cytometry and/or western blot and the functionality of expressed CARs was evaluated in vitro. Results Blocking TBK1/IKKe during transduction of NK cells enabled their efficient transduction by VSV-G LVs as judged by YFPexpression of 40–50%, with half maximal effective concentrations of 1.1 µM (MRT67307), 5 µM (BX-795) and 24.8 µM (amlexanox). Focusing on MRT67307, the authors successfully generated NK cells expressing CD19-CARs or HER2-CARs with an inducible co-stimulatory molecule. CAR NK cells exhibited increased cytolytic activity and ability to produce cytokines in comparison to untreated controls, confirming CAR functionality. Conclusions The authors demonstrate that inhibition of TBK1/IKKe enables the reliable generation of genetically modified NK cells using VSV-G LVs. The authors’ protocol can be readily adapted to generate clinical-grade NK cells and thus has the potential to facilitate the clinical evaluation of genetically modified NK cell-based therapeutics in the future.
- Published
- 2021
4. Second Allogeneic HCT Can Successfully Salvage a Subset of Patients Who Relapse Following First Allogeneic HCT: A Single-Center Retrospective Analysis of 108 Pediatric Patients
- Author
-
Dr. Rebecca Epperly, Ying Li, Subodh Selukar, Emily Zeng, Renee Madden, Swati Naik, Ewelina Mamcarz, Amr Qudeimat, Akshay Sharma, Aimee C. Talleur, Stephen Gottschalk, Ashok Srinivasan, and Brandon M. Triplett
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
5. Activity of CD19-CAR T Cell Therapy Against Extramedullary Disease in Pediatric Patients with B-ALL
- Author
-
Dr. Rebecca Epperly, Barry L. Shulkin, Asim Bag, Cheng Cheng, Swati Naik, Hiroto Inaba, Brandon M. Triplett, Stephen Gottschalk, and Aimee C. Talleur
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
6. Rewriting History: Epigenetic Reprogramming of CD8+ T Cell Differentiation to Enhance Immunotherapy
- Author
-
Stephen Gottschalk, Caitlin C. Zebley, and Ben Youngblood
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Cell Differentiation ,Immunotherapy ,Chimeric antigen receptor ,Immune checkpoint ,030104 developmental biology ,medicine.anatomical_structure ,T cell differentiation ,Cancer research ,Reprogramming ,CD8 ,030215 immunology - Abstract
The full potential of T cell-based immunotherapies remains limited by a variety of T cell extrinsic and intrinsic immunosuppressive mechanisms that can become imprinted to stably reduce the antitumor ability of T cells. Here, we discuss recent insights into memory CD8+ T cell differentiation and exhaustion and the association of these differentiation states with clinical outcomes during immune checkpoint blockade and chimeric antigen receptor (CAR) T cell therapeutic modalities. We consider the barriers limiting immunotherapy with a focus on epigenetic regulation impeding efficacy of adoptively transferred T cells and other approaches that augment T cell responses such as immune checkpoint blockade. Furthermore, we outline conceptual and technical breakthroughs that can be applied to existing therapeutic approaches and to the development of novel cutting-edge strategies.
- Published
- 2020
7. Preliminary Results from a Phase 1 Trial Showing Safety and Anti-Leukemic Activity of CD123-CAR T Cells in Pediatric Patients with AML
- Author
-
Swati Naik, Renee Madden, Amanda Lipsitt, Timothy Lockey, Jennyfer Bran, Jeffrey E Rubnitz, Jeffery Klco, Barry L. Shulkin, Sagar L Patil, Sarah S Schell, Jeoung-Eun John Park, Janice Riberdy, Na Shang, Jaquelyn Zoine, Jennifer Wallace, Katheryn Harstead, Catherine Willis, Jean-Yves Metais, Deanna Langfitt, Sheng Zhou, Salem Akel, Michael Meagher, Brandon M. Triplett, Stephen Gottschalk, and Paulina Paulina Velasquez
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
8. A tour de force provides novel insights into CD19-CAR T cell resistance mechanisms
- Author
-
Aimee C. Talleur and Stephen Gottschalk
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
9. CAR T cell therapy for solid tumors: Fatal attraction requires adhesion
- Author
-
Matthew Bell and Stephen Gottschalk
- Subjects
General Medicine - Published
- 2022
10. Evidence generation and reproducibility in cell and gene therapy research: A call to action
- Author
-
Roderic I. Pettigrew, Sarah A. Teichmann, Nancy C. Andrews, Arlene S. Bierman, Frederick R. Appelbaum, Cameron J. Turtle, Emer Cooke, John P. A. Ioannidis, Susan E. Bates, Joni L. Rutter, Marcela V. Maus, Hans Clevers, Gerhard Bauer, Marina Cavazzana, Adrian R. Krainer, Harvey V. Fineberg, Michael A. Caligiuri, Donald E. Ingber, Donald B. Kohn, George Q. Daley, Stephen Gottschalk, Jedd D. Wolchok, Victor J. Dzau, David R. Williams, Mohamed Abou-El-Enein, Lawrence S.B. Goldstein, Andre Terzic, Mark Lawler, Lee M. Ellis, Fyodor D. Urnov, Margaret A. Hamburg, Christine L. Mummery, Malcolm K. Brenner, Aris Angelis, Peter W. Marks, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Reproducibility ,QH573-671 ,business.industry ,Genetic enhancement ,MEDLINE ,MathematicsofComputing_GENERAL ,QH426-470 ,Bioinformatics ,GeneralLiterature_MISCELLANEOUS ,Call to action ,InformationSystems_GENERAL ,Editorial ,Genetics ,Medicine ,Molecular Medicine ,business ,Cytology ,Molecular Biology - Abstract
Editorial
- Published
- 2021
11. Venetoclax-Based Combination Therapy As a Bridge to Allogeneic Hematopoietic Stem Cell Transplant in Children with Relapsed/Refractory AML
- Author
-
Thomas Pfeiffer, Ying Li, Seth E Karol, Jeffrey E Rubnitz, Rebecca Epperly, Renee Madden, Ewelina Mamcarz, Esther A Obeng, Amr Qudeimat, Akshay Sharma, Ashok Srinivasan, Ali Suliman, Aimee C Talleur, Mireya Paulina Velasquez, Stephen Gottschalk, Brandon M. Triplett, and Swati Naik
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
12. Long Term Follow up for the Development of Subsequent Malignancies in Patients Treated with Genetically Modified Immune Effectors
- Author
-
David H.M. Steffin, Ibrahim N. Muhsen, Nabil M Ahmed, Meena Hegde, Olga Dakhova, Tao Wang, Jesse Wu, Stephen Gottschalk, Sarah Whittle, Premal D. Lulla, Maksim Mamonkin, Bilal Omer, Rayne Helen Rouce, Andras Heczey, Leonid S. Metelitsa, LaQuisa Hill, Carlos A. Ramos, Cliona M. Rooney, Malcolm K. Brenner, and Helen E. Heslop
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
13. Epstein-Barr Virus (EBV)-derived BARF1 encodes CD4- and CD8-restricted epitopes as targets for T-cell immunotherapy
- Author
-
Minthran C. Ngo, Cliona M. Rooney, Chrystal U. Louis, Ulrike Gerdemann, Stephen Gottschalk, Jessica D. Luu, Mamta Kalra, and Ann M. Leen
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Herpesvirus 4, Human ,Cancer Research ,Lymphoma ,medicine.medical_treatment ,Immunology ,Human leukocyte antigen ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Major histocompatibility complex ,Immunotherapy, Adoptive ,Article ,Epitope ,Viral Matrix Proteins ,Epitopes ,Viral Proteins ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,Immunology and Allergy ,Genetics (clinical) ,Transplantation ,biology ,Cell Biology ,Immunotherapy ,Epstein–Barr virus ,HEK293 Cells ,030104 developmental biology ,Epitope mapping ,Epstein-Barr Virus Nuclear Antigens ,Oncology ,030220 oncology & carcinogenesis ,Trans-Activators ,biology.protein ,Cancer research ,CD8 - Abstract
Background aims EBV type II latency tumors, such as Hodgkin lymphoma (HL), Non-Hodgkin lymphoma (NHL) and nasopharyngeal carcinoma, express a limited array of EBV antigens including Epstein-Barr nuclear antigen (EBNA)1, latent membrane protein (LMP)1, LMP2, and BamH1-A right frame 1 (BARF1). Adoptive immunotherapy for these malignancies have focused on EBNA1, LMP1 and LMP2 because little is known about the cellular immune response to BARF1. Methods To investigate whether BARF1 is a potential T-cell immunotherapy target, we determined the frequency of BARF1-specific T-cell responses in the peripheral blood of EBV-seropositive healthy donor and patients with EBV-positive malignancies, mapped epitopes and evaluated the effector function of ex vivo–generated BARF1-specific T-cell lines. Results BARF1-specific T cells were present in the peripheral blood of 12/16 (75%) EBV-positive healthy donors and 13/20 (65%) patients with EBV-positive malignancies. Ex vivo expanded BARF1-specific T-cell lines contained CD4- and CD8-positive T-cell subpopulations, and we identified 23 BARF1 peptides, which encoded major histocompatibility complex class I– and/or II–restricted epitopes. Epitope mapping identified one human leukocyte antigen (HLA)-A*02-restricted epitope that was recognized by 50% of HLA-A*02, EBV-seropositive donors and one HLA-B*15(62)-restricted epitope. Exvivo expanded BARF1-specific T cells recognized and killed autologous, EBV-transformed lymphoblastoid cell lines and partially HLA-matched EBV-positive lymphoma cell lines. Discussion BARF1 should be considered as an immunotherapy target for EBV type II (and III) latency. Targeting BARF1, in addition to EBNA1, LMP1 and LMP2, has the potential to improve the efficacy of current T-cell immunotherapy approaches for these malignancies.
- Published
- 2019
14. Sub-myeloablative Second Transplantations with Haploidentical Donors and Post-Transplant Cyclophosphamide have limited Anti-Leukemic Effects in Pediatric Patients
- Author
-
Rebecca Epperly, Aimee C Talleur, Ying Li, Sarah Schell, MaCal Tuggle, Jean-Yves Métais, Sujuan Huang, Deqing Pei, Cheng Cheng, Renee Madden, Ewelina Mamcarz, Swati Naik, Amr Qudeimat, Akshay Sharma, Ashok Srinivasan, Ali Suliman, Stephen Gottschalk, and Brandon M. Triplett
- Subjects
Transplantation ,Hematopoietic Stem Cell Transplantation ,Graft vs Host Disease ,Cell Biology ,Hematology ,Article ,Heterocyclic Compounds ,Recurrence ,Hematologic Neoplasms ,Humans ,Molecular Medicine ,Immunology and Allergy ,Child ,Cyclophosphamide - Abstract
BACKGROUND: Pediatric patients with high-risk hematologic malignancies who experience relapse after a prior allogeneic hematopoietic cell transplant (HCT) have an exceedingly poor prognosis. A second allogeneic HCT offers the potential for long-term cure but carries high risks of both subsequent relapse and HCT-related morbidity and mortality. Using haploidentical donors for HCT (haploHCT) can expand the donor pool and potentially enhance the graft-versus-leukemia effect but is accompanied by a risk of graft-versus-host disease (GVHD). OBJECTIVES: The goal of this protocol was to intensify the antileukemia effect of haploHCT for pediatric patients with hematologic malignancies that relapsed after prior allogeneic HCT, while limiting regimen-associated toxicities. STUDY DESIGN: This phase II clinical trial evaluated a sub-myeloablative preparative regimen consisting of anti-thymocyte globulin (ATG), clofarabine, cytarabine, busulfan, and cyclophosphamide, in combination with plerixafor to sensitize leukemic blasts. Participants received a mobilized peripheral blood unmanipulated haploidentical donor graft with one dose of post-transplant cyclophosphamide as GVHD prophylaxis, followed by NK cell addback. Here we report the clinical outcomes and immune reconstitution of 17 participants treated on the study, and 5 additional patients treated on similar single-patient treatment plans. RESULTS: Of the 22 participants analyzed, 12 (55%) had active disease at the time of HCT. The regimen provided robust immune reconstitution, with 21 participants (95%) experiencing neutrophil engraftment at a median of 14 days post HCT. In this high-risk population, the overall survival was 45% (95% CI: 24%–64%), with a 12-month event-free survival of 31% (95% confidence interval [CI]: 14%–51%) and cumulative incidence of relapse at 12 months of 50% (95% CI: 27%–69%). Four participants (18%) remain in remission at >5 years follow-up. Expected HCT-related organ-specific toxicities were observed, and 13 participants (59%) experienced acute and/or chronic GVHD. CONCLUSIONS: This intensified but sub-myeloablative regimen, followed by a high-dose unmanipulated haploidentical graft, post-transplant cyclophosphamide, and NK Cell infusion, resulted in adequate immune reconstitution but failed to overcome the elevated risks of relapse and treatment-related morbidity in this high-risk population.
- Published
- 2022
15. 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
16. Pediatric Patient and Caregiver Reported Symptom Burden, Anxiety, Depression, and Quality of Life during CD19-CAR T-Cell Therapy
- Author
-
Deena R Levine, Dagny M Vaughn, Lindsay Blazin, Stephen Gottschalk, Brandon M. Triplett, Justin N Baker, and Aimee C Talleur
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
17. Safety and Tolerability of Administering Leukemia-Specific Donor T Cells (mLSTs) after Allogeneic Transplant to Pediatric Patients with AML/MDS
- Author
-
Erin E Doherty, Bilal Omer, Spyridoula Vasileiou, Ayumi Watanabe, Manik Kuvalekar, Catherine Robertson, Ghadir Sasa, Tami John, John Craddock, Brian D. Friend, Baheyeldin Salem, Caridad Martinez, Robert A. Krance, Stephen Gottschalk, Bambi Grilley, Adrian P. Gee, Malcolm K. Brenner, Helen E. Heslop, Ann M. Leen, Premal D. Lulla, and Swati Naik
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
18. CD45RA Depleted T-Cell Addback and Prophylactic Blinatumomab Administration Following Tcrαβ/CD19-Depleted Haploidentical Transplantation in Pediatric Patients with High Risk Acute Leukemia
- Author
-
Swati Naik, Ying Li, Renee Madden, Ewelina Mamcarz, Ashok Srinivasan, Akshay Sharma, Aimee C Talleur, Amr Qudeimat, Ali Suliman, Rebecca Epperly, Esther A Obeng, Mireya Paulina Velasquez, Diego Hijano, Gabriela M. Marón, Jean-Yves Metais, Stephen Gottschalk, and Brandon M. Triplett
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
19. CD45RA-Depleted Haploidentical Transplantation Combined with NK Cell Addback Results in Promising Long-Term Outcomes in Pediatric Patients with High-Risk Hematologic Malignancies
- Author
-
Swati Naik, Aimee C Talleur, Ying Li, Renee Madden, Ewelina Mamcarz, Amr Qudeimat, Akshay Sharma, Ashok Srinivasan, Ali Suliman, Rebecca Epperly, Esther A Obeng, Mireya Paulina Velasquez, Diego Hijano, Gabriela M. Marón, Jean-Yves Metais, Stephen Gottschalk, and Brandon M. Triplett
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
20. 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
21. Current Allogeneic Hematopoietic Stem Cell Transplantation for Pediatric Acute Lymphocytic Leukemia: Success, Failure and Future Perspectives—A Single-Center Experience, 2008 to 2016
- Author
-
Jo Eunji, Meenakshi Hegde, Asaf D. Yanir, Caridad Martinez, Hao Liu, Ghadir Sasa, Nabil Ahmed, Bilal Omer, Kathryn Leung, Malcolm K. Brenner, Helen E. Heslop, Robert A. Krance, Swati Naik, and Stephen Gottschalk
- Subjects
Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,Lymphoblastic Leukemia ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Single Center ,History, 21st Century ,03 medical and health sciences ,Pediatric Acute Lymphocytic Leukemia ,0302 clinical medicine ,Internal medicine ,medicine ,Success failure ,Humans ,Transplantation, Homologous ,Sibling ,Child ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,Infant ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Minimal residual disease ,Haploidentical Donor ,surgical procedures, operative ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,business ,030215 immunology - Abstract
Hematopoietic stem cell transplantation (HSCT) is the only curative option for a subset of patients with high-risk or relapsed acute lymphoblastic leukemia (ALL). Given evolving practices, it is important to continually evaluate outcomes for pediatric ALL following HSCT. Outcomes after HSCT are influenced by the type of donor used as this determines the degree and method of T cell depletion used and, consequently, specific transplant-related morbidities. We retrospectively analyzed HSCT data from our center for transplants performed between January 2008 and May 2016, comparing outcomes among different donor types. One hundred and twenty-four pediatric patients underwent HSCT from a matched sibling donor (MSD; n = 48), an unrelated matched donor (UMD; n = 56), or a haploidentical donor (n = 20). We observed a similar 3-year event-free survival (EFS) for MSD recipients (of .64) and for UMD recipients (.62), but a significantly lower EFS for recipients of haploidentical transplants (.35; P = .01). Relapse was the main cause of HSCT failure and was significantly higher in the haploidentical donor group (.47 versus .19 for MSD and .24 for UMD; P = .02). Treatment-related mortality was evenly distributed among the donor groups (.17, .16, and .15 for the MSD, UMD, and haploidentical groups, respectively). Rates of infection-related mortality were lower than previously reported. Relapse is the main obstacle for successful HSCT in the contemporary era, and this effect is most evident in recipients of haploidentical donor grafts. Newer methods to improve graft-versus-leukemia effect are being evaluated and will need to be incorporated into the management of high-risk patients.
- Published
- 2018
22. Optimizing EphA2-CAR T Cells for the Adoptive Immunotherapy of Glioma
- Author
-
Felicia Cao, Brooke Prinzing, Giedre Krenciute, Zhongzhen Yi, and Stephen Gottschalk
- Subjects
0301 basic medicine ,lcsh:QH426-470 ,Brain tumor ,EphA2 ,GBM ,CAR T cells ,Article ,03 medical and health sciences ,Antigen ,Glioma ,Genetics ,Medicine ,lcsh:QH573-671 ,Molecular Biology ,lcsh:Cytology ,business.industry ,Effector ,CD28 ,medicine.disease ,EPH receptor A2 ,Chimeric antigen receptor ,3. Good health ,lcsh:Genetics ,030104 developmental biology ,Cancer research ,Molecular Medicine ,Car t cells ,business ,human activities ,brain tumor - Abstract
Glioblastoma is the most aggressive primary brain tumor in humans and is virtually incurable with conventional therapies. Chimeric antigen receptor (CAR) T cell therapy targeting the glioblastoma antigen EphA2 is an attractive approach to improve outcomes because EphA2 is expressed highly in glioblastoma but only at low levels in normal brain tissue. Building upon our previous findings in this area, we generated and evaluated a panel of EphA2-specific CARs. We demonstrate here that T cells expressing CD28.ζ and 41BB.ζ CARs with short spacers had similar effector function, resulting in potent antitumor activity. In addition, incorporating the 41BB signaling domain into CD28.ζ CARs did not improve CAR T cell function. While we could not determine functional differences between CD28.ζ, 41BB.ζ, and CD28.41BB.ζ CAR T cells, we selected CD28.ζ CAR T cells for further clinical development based on safety consideration. Keywords: GBM, CAR T cells, EphA2, brain tumor
- Published
- 2018
23. Outcomes after Allogeneic Transplant in Patients with Wiskott-Aldrich Syndrome
- Author
-
Caridad Martinez, Robert A. Krance, Nicholas L. Rider, Bilal Omer, Alexander Ngwube, William Shearer, Ghadir Sasa, Nabil Ahmed, Swati Naik, Stephen Gottschalk, Jordan S. Orange, Malcolm K. Brenner, Filiz O. Seeborg, Meng Fen Wu, Carl E. Allen, I. Celine Hanson, Helen E. Heslop, Lenora M. Noroski, Lisa R. Forbes, Sarah K. Nicholas, Hao Liu, Kathryn Leung, and Meenakshi Hegde
- Subjects
Adult ,Male ,medicine.medical_specialty ,Transplantation Conditioning ,Adolescent ,Platelet Engraftment ,Wiskott–Aldrich syndrome ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Gastroenterology ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,medicine ,Humans ,Platelet ,In patient ,Child ,Immunodeficiency ,Retrospective Studies ,Transplantation ,business.industry ,Hematopoietic Stem Cell Transplantation ,Hematology ,Allografts ,medicine.disease ,Wiskott-Aldrich Syndrome ,Survival Rate ,Child, Preschool ,030220 oncology & carcinogenesis ,Allogeneic hsct ,Transfusion dependence ,Female ,business ,Follow-Up Studies ,030215 immunology - Abstract
Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder characterized by a triad of immunodeficiency, eczema, and thrombocytopenia. Currently, hematopoietic stem cell transplantation (HSCT) is the most reliable curative treatment with excellent results for patients with HLA-matched family or unrelated donors. However, even after fully myeloablative preparative regimens, mixed donor chimerism is a potential concern. We performed a retrospective chart review of 12 children who underwent allogeneic HSCT for WAS to report our experience. The median age at transplant was 10.5 months (range, 3 to 39). The median nucleated cell dose from the marrow was 4.55 × 109/kg (range, .3 to 7.9). The median times to neutrophil and platelet engraftment were 19 days (range, 13 to 27) and 18.5 days (range, 12 to 31), respectively. The rate of overall survival was 92% with median follow-up of 67 months (range, 3 to 146). Two patients developed grade IV acute graft-versus-host disease, and 1 died on day +99. Five of 12 patient's (42%) had mixed donor chimerism (range, 12% to 85%) at day +180. None of the pretransplant patient parameters was predictive of mixed chimerism. Nonetheless, of these 5 patients, 2 had normalization of the platelet count despite the mixed chimerism, 2 had full donor chimerism after receiving a second transplant with the same donor, and 1 remains transfusion dependent awaiting a second transplant. Hence, even with a significant rate of mixed chimerism, HSCT provides substantial benefit to WAS patients, with excellent overall survival.
- Published
- 2018
24. In Situ Liver Expression of HBsAg/CD3-Bispecific Antibodies for HBV Immunotherapy
- Author
-
Robert L. Kruse, Karl-Dimiter Bissig, Xavier Legras, Frank P. Pankowicz, Mercedes Barzi, Thomas Shum, and Stephen Gottschalk
- Subjects
0301 basic medicine ,Adoptive cell transfer ,HBsAg ,lcsh:QH426-470 ,viral hepatitis ,liver ,medicine.disease_cause ,Article ,Immunoglobulin G ,03 medical and health sciences ,Antigen ,Genetics ,medicine ,lcsh:QH573-671 ,Molecular Biology ,Hepatitis B virus ,biology ,lcsh:Cytology ,T cell ,virus diseases ,cccDNA ,medicine.disease ,gene therapy ,Virology ,digestive system diseases ,3. Good health ,bispecific antibody ,lcsh:Genetics ,030104 developmental biology ,Immunology ,biology.protein ,Molecular Medicine ,immunotherapy ,Antibody ,Viral hepatitis ,hepatitis B virus - Abstract
Current therapies against hepatitis B virus (HBV) do not reliably cure chronic infection, necessitating new therapeutic approaches. The T cell response can clear HBV during acute infection, and the adoptive transfer of antiviral T cells during bone marrow transplantation can cure patients of chronic HBV infection. To redirect T cells to HBV-infected hepatocytes, we delivered plasmids encoding bispecific antibodies directed against the viral surface antigen (HBsAg) and CD3, expressed on almost all T cells, directly into the liver using hydrodynamic tail vein injection. We found a significant reduction in HBV-driven reporter gene expression (184-fold) in a mouse model of acute infection, which was 30-fold lower than an antibody only recognizing HBsAg. While bispecific antibodies triggered, in part, antigen-independent T cell activation, antibody production within hepatocytes was non-cytotoxic. We next tested the bispecific antibodies in a different HBV mouse model, which closely mimics the transcriptional template for HBV, covalently closed circular DNA (cccDNA). We found that the antiviral effect was noncytopathic, mediating a 495-fold reduction in HBsAg levels at day 4. At day 33, bispecific antibody-treated mice exhibited 35-fold higher host HBsAg immunoglobulin G (IgG) antibody production versus untreated groups. Thus, gene therapy with HBsAg/CD3-bispecific antibodies represents a promising therapeutic strategy for patients with HBV.
- Published
- 2017
25. Development of a cgmp-compliant process to manufacture donor-derived, CD45RA-depleted memory cd19- car T-Cells
- Author
-
Aimee C Talleur, Salem M. Akel, N. Shang, Paulina Velasquez, J. Park, Stephen Gottschalk, J. Moore, S. Zhou, F. Zheng, Janice M. Riberdy, Jean-Yves Metais, Y. Kim Hoehamer, and Brandon M. Triplett
- Subjects
Cancer Research ,Transplantation ,biology ,Chemistry ,Immunology ,Cell Biology ,CD19 ,Cell biology ,Oncology ,biology.protein ,Immunology and Allergy ,Donor derived ,Car t cells ,Process (anatomy) ,Genetics (clinical) - Published
- 2021
26. T Cell-Activating Mesenchymal Stem Cells as a Biotherapeutic for HCC
- Author
-
Mireya Paulina Velasquez, Abishek Vaidya, Stephen Gottschalk, Andras Heczey, Daniel L. Galvan, David Torres, Arpad Szoor, Zhuyong Mei, and Adrian P. Gee
- Subjects
0301 basic medicine ,Cancer Research ,CD3 ,T cell ,medicine.medical_treatment ,lcsh:RC254-282 ,Viral vector ,03 medical and health sciences ,Antigen ,medicine ,Pharmacology (medical) ,Elméleti orvostudományok ,biology ,Mesenchymal stem cell ,hepatocellular carcinoma ,Orvostudományok ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,Genetically modified organism ,GPC3 ,bispecific antibody ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Immunology ,biology.protein ,Cancer research ,Molecular Medicine ,Original Article ,immunotherapy ,CD80 - Abstract
The outcome for advanced stage hepatocellular carcinoma (HCC) remains poor, highlighting the need for novel therapies. Genetically modified mesenchymal stem cells (MSCs) are actively being explored as cancer therapeutics due to their inherent ability to migrate to tumor sites. We reasoned that MSCs can be genetically modified to redirect T cells to Glypican-3 (GPC3)+ HCC, and genetically modified these with viral vectors encoding a GPC3/CD3 bispecific T cell engager (GPC3-ENG), a bispecifc T cell engager specific for an irrelevant antigen (EGFRvIII), and/or costimulatory molecules (CD80 and 41BBL). Coculture of GPC3+ cells, GPC3-ENG MSCs, and T cells resulted in T cell activation, as judged by interferon γ (IFNγ) production and killing of tumor cells by T cells. Modification of GPC3-ENG MSCs with CD80 and 41BBL was required for antigen-dependent interleukin-2 (IL-2) production by T cells and resulted in faster tumor cell killing by redirected T cells. In vivo, GPC3-ENG MSCs ± costimulatory molecules had antitumor activity in the HUH7 HCC xenograft model, resulting in a survival advantage. In conclusion, MSCs genetically modified to express GPC3-ENG ± costimulatory molecules redirect T cells to GPC3+ tumor cells and have potent antitumor activity. Thus, further preclinical exploration of our modified approach to GPC3-targeted immunotherapy for HCC is warranted., Graphical Abstract
- Published
- 2017
27. Second Allogeneic Hematopoietic Cell Transplant Is a Successful Salvage Modality for Pediatric Patients Who Relapse after First Transplant
- Author
-
Amr Qudeimat, Cheng Cheng, Renee Madden, Ashok Srinivasan, Sujuan Huang, Akshay Sharma, Aimee C Talleur, Ying Li, Stephen Gottschalk, Ewelina Mamcarz, and Brandon M. Triplett
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Chemotherapy ,Hematopoietic cell ,business.industry ,medicine.medical_treatment ,Medical record ,Retrospective cohort study ,Hematology ,Disease ,Logistic regression ,Donor lymphocyte infusion ,surgical procedures, operative ,hemic and lymphatic diseases ,Internal medicine ,medicine ,business - Abstract
Introduction Disease relapse remains a major cause of treatment failure in pediatric patients receiving allogeneic hematopoietic cell transplantation (HCT) for high-risk acute leukemias (AL) or myelodysplastic syndrome (MDS). Comprehensive data on prognostic factors and outcomes after post-transplant relapse in pediatric patients is lacking. Objective The goal of this study was to identify factors associated with survival following post-transplant relapse. Methods We performed a retrospective study of pediatric patients who experienced disease relapse following their 1st HCT for AL or MDS between 1990 – 2018 at our institution. Patient, disease and transplant-related characteristics were extracted from the medical record. Overall survival (OS) was estimated by the Kaplan-Meier (KM) method and compared using the log-rank test. Cox and logistic regression were performed to identify factors statistically associated with outcomes after post-transplant relapse. Results During the study period, 703 patients received 1st HCT, of which 221 (31%) relapsed at a median of 3.9 months after HCT. Of these relapsed patients, 60% (n = 132) received some chemotherapy or supportive care after post-transplant relapse, 13% (n = 28) received donor lymphocyte infusion only and 28% (n = 61) were able to proceed to a 2nd HCT. Patients who received a 2nd HCT were more likely to have been in first remission at the time of 1st HCT (P = 0.02), experienced relapse more than 6 months after 1st HCT (P Conclusion Duration of remission before post-transplant relapse was identified as the strongest independent predictor of a patient's ability to receive a 2nd HCT as well as survival after relapse. The 3-year OS for patients who received a 2nd HCT was 6 times higher than that for patients who did not receive a 2nd HCT. Although the outcomes after 2nd HCT are not commendable, they are improving with time. Our data supports the argument that, when feasible, pediatric patients experiencing post-transplant relapse should be considered for 2nd HCT.
- Published
- 2020
28. Long term follow up of subsequent malignancies in patients treated with genetically modified immune effectors
- Author
-
Rayne H. Rouce, Helen E. Heslop, David H.M. Steffin, Cliona M. Rooney, T. Wang, Ibrahim N. Muhsen, Meenakshi Hegde, Olga Dakhova, Stephen Gottschalk, S. Whittle, Malcolm K. Brenner, Premal Lulla, LaQuisa Hill, Bilal Omer, Leonid S. Metelitsa, Carlos A. Ramos, Maksim Mamonkin, Meng-Fen Wu, Andras Heczey, and Nabil Ahmed
- Subjects
Cancer Research ,Transplantation ,Long term follow up ,business.industry ,Effector ,Immunology ,Cell Biology ,Genetically modified organism ,Immune system ,Oncology ,Immunology and Allergy ,Medicine ,In patient ,business ,Genetics (clinical) - Published
- 2021
29. A facile radiosynthesis of [18F]FBA-Pep42, a cyclic peptide targeting glucose-regulated protein 78 (GRP78)
- Author
-
Stephen Gottschalk, Amy Vavere, Hao Yang, Paulina Velasquez, and Nikhil Hebbar
- Subjects
chemistry.chemical_classification ,Cancer Research ,biology ,chemistry ,Biochemistry ,Glucose-regulated protein ,Radiosynthesis ,biology.protein ,Molecular Medicine ,Radiology, Nuclear Medicine and imaging ,Cyclic peptide - Published
- 2021
30. Successful SCID gene therapy in infant with disseminated BCG
- Author
-
Stephen Gottschalk, Harry L. Malech, Michael D. Neel, Armita Bahrami, Gabriela Maron, Jennifer M. Puck, Ewelina Mamcarz, Morton J. Cowan, and Sue C. Kaste
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Genetic enhancement ,MEDLINE ,Infant ,Genetic Therapy ,Mycobacterium bovis ,Article ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Severe Combined Immunodeficiency ,business - Published
- 2021
31. Haploidentical CD45RA-Negative Donor Lymphocyte Infusions Are Feasible, Safe and Associated with Clinical Benefit
- Author
-
Ying Li, Salem M. Akel, Ashok Srinivasan, Aimee C Talleur, Amr Qudeimat, Brandon M. Triplett, Ewelina Mamcarz, Stephen Gottschalk, Akshay Sharma, and Renee Madden
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Total blood ,business.industry ,T cell ,Lymphocyte ,Hematology ,Leukapheresis ,medicine.disease ,03 medical and health sciences ,Leukemia ,0302 clinical medicine ,medicine.anatomical_structure ,immune system diseases ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Dose escalation ,Dosing ,business ,030215 immunology - Abstract
Introduction Donor T-cell reconstitution following hematopoietic cell transplantation (HCT) is critical for infection and leukemic control. Donor lymphocyte infusions (DLI) provide additional donor T cells, but may cause graft-versus-host-disease (GVHD) from alloreactive T-cells. As GVHD risk increases with increasing HLA-mismatch, conventional haploidentical DLI doses are often limited to 1 × 105 T cells/kg. Since the majority of alloreactive T cells reside in the naive (CD45RA-positive) subset, CD45RA-depleted DLI may provide a diverse lymphocyte repertoire to aid in infection and leukemic disease control, with decreased GVHD risk. Herein, we report on feasibility, safety, and early efficacy of escalating doses of haploidentical memory DLI. Methods Donors underwent leukapheresis without mobilization; 2 total blood volumes were collected. Depletion of CD45RA-positive cells was performed using the CliniMACS device (Miltenyi Biotec). DLI dosing typically started at 1 × 105 T cells/kg, with dose escalation every 4 weeks based on clinical need. Results All donors collected successfully in one attempt. Following CD45RA-depletion, median residual T cell dose was 45 × 106/kg and CD45RA-positive T cell dose was 3.9 log depletion), allowing multiple doses to be stored for future use (Figure 1). Twelve patients received a total of 29 DLIs (median infusions=2, range 1–5; Figure 2). Indications for DLI included detectible leukemia (6), mixed chimerism (6), and lymphopenia/viral infection (3); 3 recipients had multiple indications. First DLI was infused a median of 101.5 days post-HCT (range 41-356). Twenty-three doses had no detectible CD45RA-positive T cells ( Discussion CD45RA-depletion is feasible and effective in removing naive donor T-cells. Haploidentical CD45RA-depleted DLIs were well tolerated and associated with low incidence of acute GVHD, even with T-cell doses as high as 10 × 106/kg. The majority of patients showed clinical benefit from DLI, with the exception of leukemia disease control. We are currently further investigating the efficacy and safety of CD45RA-depleted DLI as part of a prospective clinical haploidentical HCT trial.
- Published
- 2020
32. Adoptive T-Cell Therapy for Acute Lymphoblastic Leukemia Targeting Multiple Tumor-Associated Antigens
- Author
-
Ann M. Leen, Cliona M. Rooney, Juan F. Vera, Swati Naik, Stephen Gottschalk, Premal Lulla, Robert A. Krance, Ifigeneia Tzannou, Adrian P. Gee, and Malcolm K. Brenner
- Subjects
Transplantation ,PRAME ,Adoptive cell transfer ,biology ,business.industry ,ELISPOT ,T cell ,Hematology ,Epitope ,CD19 ,medicine.anatomical_structure ,Antigen ,Immunology ,medicine ,biology.protein ,business ,CD8 - Abstract
Introduction HSCT is a curative option for patients with high-risk ALL but relapse remains the major cause of treatment failure. CD19 CAR T cells have shown remarkable efficacy in treating leukemic relapse post-HSCT but their use is limited to CD19+ malignancies and antigen negative relapses are increasingly being reported. To overcome these limitations, we developed a strategy to generate donor-derived T cell lines simultaneously targeting PRAME, WT1 and Survivin (multiTAAs) expressed by both B and T ALL for adoptive transfer to high risk HSCT recipients. Methods We generated donor multiTAA-T cells by culturing PBMCs with autologous DCs loaded with a mastermix of pepmixes spanning all 3 target antigens in the presence of a Th1-polarizing/pro-proliferative cytokine cocktail. Results To date, we have generated 14 clinical multiTAA-T cell lines comprising CD3+ T cells (mean 94±9%) with a mixture of CD4+ (mean 21±28%) and CD8+ (mean 52±24 %) cells, which expressed central and effector memory markers and recognized the targeted antigens based on IFNg ELIspot analysis. None of the lines reacted against non-malignant patient-derived cells (4±3% specific lysis; E:T 20:1) - a study release criterion. Thus far we have treated 10 high-risk ALL patients to prevent disease relapse post-transplant (Table 1). Infusions were well tolerated with no dose-limiting toxicity, GVHD, CRS or other adverse events. Two patients were not evaluable per study criteria as they received >0.5mg/kg of steroids within 4 weeks of infusion and were replaced. Seven of the 8 remaining patients infused remain in CCR a median of 9 months post-infusion (range 1-23 months). We detected the expansion of tumor-reactive T cells in patient 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. The single patient who relapsed showed no evidence of tumor-directed T cell expansion despite receiving 3 additional infusions at 4 week intervals. Conclusion Infusion of donor-derived multiTAA-T cells to patients with ALL post-HSCT is feasible, safe and as evidenced by expansion and antigen spreading in patients, may contribute to disease control. This strategy maybe a promising approach to prevent leukemic relapse after HSCT.
- Published
- 2019
33. Outcomes after Second Hematopoietic Stem Cell Transplantations in Pediatric Patients with Relapsed Hematological Malignancies
- Author
-
Kathryn Leung, Ngoc Yen Nguyen, Meng Fen Wu, Caridad Martinez, Malcolm K. Brenner, Robert A. Krance, Helen E. Heslop, Swati Naik, Stephen Gottschalk, and Ghadir Sasa
- Subjects
Male ,Risk ,Oncology ,medicine.medical_specialty ,Prognostic variable ,Transplantation Conditioning ,Adolescent ,medicine.medical_treatment ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Hematological malignancies ,Young Adult ,Recurrence ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Remission status ,Relapse ,Child ,Survival analysis ,Retrospective Studies ,Cause of death ,Pediatric ,Transplantation ,Second transplantation ,business.industry ,Siblings ,Hazard ratio ,Infant ,Retrospective cohort study ,Hematology ,Myeloablative Agonists ,Survival Analysis ,Surgery ,Treatment Outcome ,Child, Preschool ,Hematologic Neoplasms ,Chronic Disease ,Cohort ,Female ,Unrelated Donors ,business ,Immunosuppressive Agents - Abstract
Relapse of hematological malignancies after hematopoietic stem cell transplantation (HCT) is associated with poor prognosis. A second HCT represents one of the few therapeutic options for these high-risk patients. For children undergoing second HCT, the outcome data are particularly limited. We, therefore, conducted a retrospective single-institution study and report the outcomes and prognostic variables associated with overall survival (OS) and relapse in 43 pediatric patients who underwent a second HCT between 2000 and 2013. Eleven of the 43 patients who underwent transplantation remain alive and disease-free at a median follow-up of 49 months (range, 5 to 127 months). The 5-year probability of OS for the entire cohort was 24%. Patients who had early relapse (6 months), with 5-year OS at 11% versus 34%, respectively (hazard ratio [HR], 2.24; 95% confidence interval [CI], 1.21 to 4.93; P = .013). Active disease at time of second HCT was also associated with a significantly increased risk of relapse (subdistribution hazard ratio [SHR], 2.36; P = .049) for the entire cohort and relapse was the most frequent cause of death (23 of 32; 72%). On subgroup analysis for the 34 patients with leukemia alone, presence of active disease was associated both with a significant decrease in OS (SHR, 2.28; 95% CI, 1.02 to 5.09; P = .044) and significant increase in the rate of relapse (SHR, 2.46; P = .046). By contrast, underlying disease, donor source, conditioning regimen, or development of GVHD did not modify OS or rate of relapse. Hence, a second HCT appears to be a useful therapeutic option in children with relapsed hematological malignancies that is most likely to benefit those individuals with late onset of relapse and with low disease burden at the time of transplantation.
- Published
- 2015
- Full Text
- View/download PDF
34. T-Cell Immunotherapy: Looking Forward
- Author
-
Stephen Gottschalk, David Baltimore, Carl H. June, Linda Gargiulo, Michael C. Jensen, Jacqueline Corrigan-Curay, Steven A. Rosenberg, Eugene Rosenthal, Crystal L. Mackall, Philip D. Greenberg, Antoni Ribas, Brian G. Till, Renier J. Brentjens, Marina O'Reilly, Helen E. Heslop, Richard P. Junghans, Maureen Montgomery, Michel Sadelain, Daniel J. Powell, Laurence J.N. Cooper, Donald B. Kohn, Stephen J. Forman, Hans-Peter Kiem, Robert Jambou, Amy P. Patterson, and Oliver W. Press
- Subjects
Research design ,Pharmacology ,education.field_of_study ,Process management ,Best practice ,Population ,Genetic therapy ,3. Good health ,Immunology ,Drug Discovery ,Genetics ,Molecular Medicine ,Business ,education ,T cell immunotherapy ,Molecular Biology - Abstract
The rapidly expanding field of T-cell immunotherapy has experienced clinical successes along with some serious toxicities. “T Cell Immunotherapy: Optimizing Trial Design,” a workshop sponsored by the National Institutes of Health’s (NIH’s) Office of Biotechnology Activities (OBA), brought together researchers to discuss the scientific advances and share new data on key trial design issues, including the selection of new targets, optimizing the T-cell population, preconditioning regimens, strategies to promote persistence of cells, and analysis and management of acute reactions to T-cell infusions with the goal of identifying best practices and a research agenda that will facilitate further development and maximize the safety of this promising approach.
- Published
- 2014
- Full Text
- View/download PDF
35. T cells redirected to interleukin-13Rα2 with interleukin-13 mutein–chimeric antigen receptors have anti-glioma activity but also recognize interleukin-13Rα1
- Author
-
Tania Rodriguez-Cruz, Zhongzhen Yi, Claudia Gerken, Stephen Gottschalk, Meenakshi Hegde, Simone Krebs, Nabil Ahmed, and Kevin Chow
- Subjects
Cancer Research ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Biology ,Mice ,Interleukin 21 ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Genetics (clinical) ,Interleukin 3 ,Transplantation ,Immunotoxins ,Interleukin ,Cell Biology ,Interleukin-13 Receptor alpha1 Subunit ,Xenograft Model Antitumor Assays ,Molecular biology ,Recombinant Proteins ,Chimeric antigen receptor ,Gene Expression Regulation, Neoplastic ,Amino Acid Substitution ,Oncology ,Interleukin 13 ,Interleukin-13 Receptor alpha2 Subunit ,Immunotherapy ,Glioblastoma - Abstract
Background aims Outcomes for patients with glioblastoma remain poor despite aggressive multimodal therapy. Immunotherapy with genetically modified T cells expressing chimeric antigen receptors (CARs) targeting interleukin (IL)13Rα2, human epidermal growth factor receptor 2, epidermal growth factor variant III or erythropoietin-producing hepatocellular carcinoma A2 has shown promise for the treatment of glioma in preclinical models. On the basis of IL13Rα2 immunotoxins that contain IL13 molecules with one or two amino acid substitutions (IL13 muteins) to confer specificity to IL13Rα2, investigators have constructed CARS with IL13 muteins as antigen-binding domains. Whereas the specificity of IL13 muteins in the context of immunotoxins is well characterized, limited information is available for CAR T cells. Methods We constructed four second-generation CARs with IL13 muteins with one or two amino acid substitutions, and evaluated the effector function of IL13-mutein CAR T cells in vitro and in vivo . Results T cells expressing all four CARs recognized IL13Rα1 or IL13Rα2 recombinant protein in contrast to control protein (IL4R) as judged by interferon-γ production. IL13 protein produced significantly more IL2, indicating that IL13 mutein–CAR T cells have a higher affinity to IL13Rα2 than to IL13Rα1. In cytotoxicity assays, CAR T cells killed IL13Rα1- and/or IL13Rα2-positive cells in contrast to IL13Rα1- and IL13Rα2-negative controls. Although we observed no significant differences between IL13 mutein–CAR T cells in vitro , only T cells expressing IL13 mutein–CARs with an E13K amino acid substitution had anti-tumor activity in vivo that resulted in a survival advantage of treated animals. Conclusions Our study highlights that the specificity/avidity of ligands is context-dependent and that evaluating CAR T cells in preclinical animal model is critical to assess their potential benefit.
- Published
- 2014
36. T-cell Engager-armed Oncolytic Vaccinia Virus Significantly Enhances Antitumor Therapy
- Author
-
Xingbing Wang, Feng Yu, Xiao-Tong Song, David L. Bartlett, Z. Sheng Guo, and Stephen Gottschalk
- Subjects
Pharmacology ,CD3 ,T cell ,Biology ,Virology ,Virus ,3. Good health ,Oncolytic virus ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Antigen ,Cell culture ,Drug Discovery ,biology.protein ,medicine ,Genetics ,Oncolytic Virus Therapy ,Molecular Medicine ,Vaccinia ,Molecular Biology - Abstract
Oncolytic vaccinia virus (VV) therapy has shown promise in preclinical models and in clinical studies. However, complete responses have rarely been observed. This lack of efficacy is most likely due to suboptimal virus spread through the tumor resulting in limited tumor cell destruction. We reasoned that redirecting T cells to the tumor has the potential to improve the antitumor activity of oncolytic VVs. We, therefore, constructed a VV encoding a secretory bispecific T-cell engager consisting of two single- chain variable fragments specific for CD3 and the tumor cell surface antigen EphA2 (EphA2-T-cell engager-armed VV (EphA2-TEA-VV)). In vitro, EphA2-TEA-VV's ability to replicate and induce oncolysis was similar to that of unmodified virus. However, only tumor cells infected with EphA2-TEA-VV induced T-cell activation as judged by the secretion of interferon-γ and interleukin-2. In coculture assays, EphA2-TEA-VV not only killed infected tumor cells, but in the presence of T cells, it also induced bystander killing of noninfected tumor cells. In vivo, EphA2-TEA-VV plus T cells had potent antitumor activity in comparison with control VV plus T cells in a lung cancer xenograft model. Thus, arming oncolytic VVs with T-cell engagers may represent a promising approach to improve oncolytic virus therapy.
- Published
- 2014
- Full Text
- View/download PDF
37. Autologous CD19-CAR T-Cells for the Treatment of Acute Lymphoblastic Leukemia in Pediatric and Young Adult Patients: An initial Report from an Institutional Phase I/II Study
- Author
-
Brandon M. Triplett, Deanna Langfitt, Sheng Zhou, Wenting Zheng, Byoung Y. Ryu, Paulina Velasquez, Alisha Gaboriault, Jeremy Chase Crawford, Robert E. Throm, Timothy D. Lockey, Janice M. Riberdy, Guolian Kang, Clifford A. Froelich, Terrence L. Geiger, Aimee C Talleur, Michael M Meagher, Benjamin Youngblood, Jean-Yves Metais, Amr Qudeimat, Abishek Vaidya, Stephen Gottschalk, Catherine Willis, and Paul G. Thomas
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Lymphoblastic Leukemia ,Hematology ,CD19 ,Phase i ii ,Internal medicine ,medicine ,biology.protein ,Young adult ,Car t cells ,business - Published
- 2019
38. Safety and clinical efficacy of rapidly-generated trivirus-directed T cells as treatment for adenovirus, EBV, and CMV infections after allogeneic hematopoietic stem cell transplant
- Author
-
Cliona M. Rooney, Usha L. Katari, Ann M. Leen, Kathryn Leung, Ulrike Gerdemann, John Craddock, Hao Liu, Helen E. Heslop, Anastasia Papadopoulou, Stephen Gottschalk, Jacqueline M. Keirnan, Alana A. Kennedy-Nasser, Robert A. Krance, Caridad Martinez, and Malcolm K. Brenner
- Subjects
Male ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Adoptive cell transfer ,Adolescent ,Adenoviridae Infections ,viruses ,medicine.medical_treatment ,T cell ,Cytomegalovirus ,Hematopoietic stem cell transplantation ,Biology ,medicine.disease_cause ,Virus ,Adenoviridae ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Genetics ,medicine ,Humans ,Transplantation, Homologous ,Child ,Antigens, Viral ,Molecular Biology ,Epstein–Barr virus infection ,030304 developmental biology ,Pharmacology ,0303 health sciences ,DNA Viruses ,Hematopoietic Stem Cell Transplantation ,virus diseases ,Herpesviridae Infections ,medicine.disease ,Adoptive Transfer ,Virology ,3. Good health ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,Cytomegalovirus Infections ,Immunology ,Molecular Medicine ,Original Article ,Female ,Viral load ,T-Lymphocytes, Cytotoxic - Abstract
Adoptive transfer of virus-specific T cells can prevent and treat serious infections with Epstein-Barr virus (EBV), cytomegalovirus (CMV), and adenovirus (Adv) after allogeneic hematopoietic stem cell transplant. It has, however, proved difficult to make this approach widely available since infectious virus and viral vectors are required for T cell activation, followed by an intensive and prolonged culture period extending over several months. We now show that T cells targeting a range of viral antigens derived from EBV, CMV, and Adv can be reproducibly generated in a single culture over a 2-3-week period, using methods that exclude all viral components and employ a much-simplified culture technology. When administered to recipients of haploidentical (n = 5), matched unrelated (n = 3), mismatched unrelated (n = 1) or matched related (n = 1) transplants with active CMV (n = 3), Adv (n = 1), EBV (n = 2), EBV+Adv (n = 2) or CMV+Adv (n = 2) infections, the cells produced complete virological responses in 80%, including all patients with dual infections. In each case, a decrease in viral load correlated with an increase in the frequency of T cells directed against the infecting virus(es); both immediate and delayed toxicities were absent. This approach should increase both the feasibility and applicability of T cell therapy. The trial was registered at www.clinicaltrials.gov as NCT01070797.
- Published
- 2013
39. Combinational Targeting Offsets Antigen Escape and Enhances Effector Functions of Adoptively Transferred T Cells in Glioblastoma
- Author
-
Meenakshi, Hegde, Amanda, Corder, Kevin K H, Chow, Malini, Mukherjee, Aidin, Ashoori, Yvonne, Kew, Yi Jonathan, Zhang, David S, Baskin, Fatima A, Merchant, Vita S, Brawley, Tiara T, Byrd, Simone, Krebs, Meng Fen, Wu, Hao, Liu, Helen E, Heslop, Stephen, Gottschalk, Stephen, Gottachalk, Eric, Yvon, and Nabil, Ahmed
- Subjects
Adoptive cell transfer ,Receptor, ErbB-2 ,Recombinant Fusion Proteins ,T-Lymphocytes ,T cell ,Receptors, Antigen, T-Cell ,Mice, SCID ,Streptamer ,Biology ,Models, Biological ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,Cell Line, Tumor ,Drug Discovery ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Antigen-presenting cell ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Adoptive Transfer ,Combined Modality Therapy ,Xenograft Model Antitumor Assays ,Molecular biology ,Chimeric antigen receptor ,3. Good health ,HEK293 Cells ,medicine.anatomical_structure ,Tumor Escape ,030220 oncology & carcinogenesis ,Interleukin-13 Receptor alpha2 Subunit ,Cancer research ,Molecular Medicine ,Original Article ,Glioblastoma - Abstract
Preclinical and early clinical studies have demonstrated that chimeric antigen receptor (CAR)-redirected T cells are highly promising in cancer therapy. We observed that targeting HER2 in a glioblastoma (GBM) cell line results in the emergence of HER2-null tumor cells that maintain the expression of nontargeted tumor-associated antigens. Combinational targeting of these tumor-associated antigens could therefore offset this escape mechanism. We studied the single-cell coexpression patterns of HER2, IL-13Rα2, and EphA2 in primary GBM samples using multicolor flow cytometry and immunofluorescence, and applied a binomial routine to the permutations of antigen expression and the related odds of complete tumor elimination. This mathematical model demonstrated that cotargeting HER2 and IL-13Rα2 could maximally expand the therapeutic reach of the T cell product in all primary tumors studied. Targeting a third antigen did not predict an added advantage in the tumor cohort studied. We therefore generated bispecific T cell products from healthy donors and from GBM patients by pooling T cells individually expressing HER2 and IL-13Rα2-specific CARs and by making individual T cells to coexpress both molecules. Both HER2/IL-13Rα2-bispecific T cell products offset antigen escape, producing enhanced effector activity in vitro immunoassays (against autologous glioma cells in the case of GBM patient products) and in an orthotopic xenogeneic murine model. Further, T cells coexpressing HER2 and IL-13Rα2-CARs exhibited accentuated yet antigen-dependent downstream signaling and a particularly enhanced antitumor activity.
- Published
- 2013
40. Antitumor Effects of Chimeric Receptor Engineered Human T Cells Directed to Tumor Stroma
- Author
-
Sunitha Kakarla, Lisa L. Wang, Hao Liu, Klaus Pfizenmaier, Melinda Mata, Stephen Gottschalk, David R. Rowley, Donald R. Shaffer, Meng-Fen Wu, Kevin Chow, and Xiao-Tong Song
- Subjects
Cytotoxicity, Immunologic ,Male ,Adoptive cell transfer ,congenital, hereditary, and neonatal diseases and abnormalities ,Stromal cell ,medicine.medical_treatment ,Recombinant Fusion Proteins ,T-Lymphocytes ,Genetic Vectors ,Gene Expression ,Biology ,Interleukin 21 ,Mice ,Antigen ,Cell Line, Tumor ,Neoplasms ,Endopeptidases ,Gene Order ,Drug Discovery ,medicine ,Genetics ,Cytotoxic T cell ,Animals ,Humans ,IL-2 receptor ,Lung ,Molecular Biology ,Pharmacology ,Serine Endopeptidases ,Membrane Proteins ,Immunotherapy ,Fibroblasts ,Molecular biology ,digestive system diseases ,Disease Models, Animal ,Receptors, Antigen ,Gelatinases ,Cancer cell ,Cancer research ,Cytokines ,Molecular Medicine ,Original Article ,Inflammation Mediators - Abstract
Cancer-associated fibroblasts (CAFs), the principle component of the tumor-associated stroma, form a highly protumorigenic and immunosuppressive microenvironment that mediates therapeutic resistance. Co-targeting CAFs in addition to cancer cells may therefore augment the antitumor response. Fibroblast activation protein-α (FAP), a type 2 dipeptidyl peptidase, is expressed on CAFs in a majority of solid tumors making it an attractive immunotherapeutic target. To target FAP-positive CAFs in the tumor-associated stroma, we genetically modified T cells to express a FAP-specific chimeric antigen receptor (CAR). The resulting FAP-specific T cells recognized and killed FAP-positive target cells as determined by proinflammatory cytokine release and target cell lysis. In an established A549 lung cancer model, adoptive transfer of FAP-specific T cells significantly reduced FAP-positive stromal cells, with a concomitant decrease in tumor growth. Combining these FAP-specific T cells with T cells that targeted the EphA2 antigen on the A549 cancer cells themselves significantly enhanced overall antitumor activity and conferred a survival advantage compared to either alone. Our study underscores the value of co-targeting both CAFs and cancer cells to increase the benefits of T-cell immunotherapy for solid tumors.
- Published
- 2013
- Full Text
- View/download PDF
41. IVIG Prophylaxis in Pediatric Patients Undergoing Hematopoietic Stem Cell Transplant: A Retrospective Analysis of Monthly Intravenous Immunoglobulin Infusion vs. IgG Level Based Dosing
- Author
-
Bilal Omer, Jennifer Foster, Swati Naik, Carl E. Allen, Malcolm K. Brenner, Ngoc-Yen Nguyen, Ghadir Sasa, Ann M. Leen, Nabil Ahmed, Stacey Shubert, Caridad Martinez, Meena Hegde, Helen E. Heslop, Stephen Gottschalk, W. Susan Cheng, Kathryn Leung, and Robert A. Krance
- Subjects
medicine.medical_specialty ,Transplantation ,biology ,business.industry ,Hematopoietic stem cell ,Hematology ,Surgery ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Retrospective analysis ,Dosing ,Antibody ,business - Published
- 2016
- Full Text
- View/download PDF
42. Allogeneic Stem Cell Transplantation in a Pediatric Patient with Whim Syndrome
- Author
-
Swati Naik, Carl E. Allen, Donald H. Mahoney, Meena Hegde, Saleh Bhar, Khaled Yassine, Kathryn S. Leung, Helen E. Heslop, Ghadir Sasa, Caridad Martinez, Robert A. Krance, Nabil Ahmed, and Stephen Gottschalk
- Subjects
Myelokathexis ,medicine.medical_specialty ,Transplantation ,Platelet Engraftment ,business.industry ,Hematology ,medicine.disease ,Gastroenterology ,Pancytopenia ,Granulocyte colony-stimulating factor ,Internal medicine ,Medicine ,Alemtuzumab ,business ,Congenital Neutropenia ,WHIM syndrome ,medicine.drug - Abstract
WHIM syndrome (warts, hypogammaglobulinemia, infections, and myelokathexis) is a rare autosomal dominant immunodeficiency disorder that is caused by a gain in function in cysteine-X-cysteine chemokine receptor 4 ( CXCR4 ) gene. Affected individuals have recurrent infections of the respiratory tract and soft tissues, and marked susceptibility to warts caused by human papilloma viruses (HPV). Patients have an increased risk of malignancy (lymphoma) and HPV-associated cancers. The optimal therapy for WHIM syndrome has not been defined and treatment has been with supportive care using intravenous immunoglobulin (IVIG) and granulocyte colony stimulating factor (G-CSF). New CXCR4 antagonist therapies are still in either phase I trials (plerixafor) or considered experimental (Chlacone-4). A successful umbilical cord blood transplant has been reported by Kriven et al . Here we describe a successful matched unrelated allogeneic stem cell transplant (SCT) in a girl with WHIM syndrome caused by a known mutation in the CXCR4 gene. This patient was diagnosed at birth with familial congenital neutropenia as findings were similar to those for the patient9s mother and maternal grandmother. A complete blood count showed pancytopenia (WBC= 1.03 x10 3 /µL, hemoglobin= 9.2 g/dL, platelet count= 74,000/µL, ANC= 70). Despite G-CSF and IVIG therapy, the patient had recurrent infections with sinusitis and pneumonia, along with progressive organomegaly with progressive pancytopenia. At the age of 4, the patient underwent matched unrelated SCT with a fully ablative regimen [busulfan 0.8 mg/kg/dose for 16 doses (days -9, -8, -7, and -6), cyclophosphamide 50 mg/kg/dose for 4 doses (days -5, -4, -3, and -2) and alemtuzumab 5 mg/day (weight-based dosage) for 3 doses (days -5, -4, and -3)]. Tacrolimus and mini methotrexate (on days +1, +3, +6 and +11) were given for GVHD prophylaxis. Neutrophil and platelet engraftment occurred on days + 21 and + 45, respectively. The patient is 100% donor chimeric. Post-transplant course has been complicated by grade II skin GVHD treated successfully with oral and topical steroids. The patient also developed adenovirus, BK virus and HHV-6 viral reactivations but without disease. The patient is now approximately 2 years post SCT with durable engraftment, normal humoral immune reconstitution and no chronic GVHD. Thus, stem cell transplant following myeloablative conditioning can be accomplished for patients with WHIM syndrome. Furthermore, transplantation performed at the patient9s young age likely prevented complications of recurrent infections and warts. Disclosures Allen: NovImmune: Consultancy, Other: unpaid; Roche: Consultancy, Other: unpaid. Heslop: Cell Medica: Other: Licensing Agreement; Celgene: Other: Collaborative research agreement.
- Published
- 2016
- Full Text
- View/download PDF
43. Rapidly Generated Multivirus-specific Cytotoxic T Lymphocytes for the Prophylaxis and Treatment of Viral Infections
- Author
-
Ulrike Gerdemann, Stephen Gottschalk, Serena K. Perna, Ann M. Leen, Usha L. Katari, Jacqueline M. Keirnan, Leslie E. Huye, Malcolm K. Brenner, Ryu Yanagisawa, Cliona M. Rooney, Helen E. Heslop, Sravya Ennamuri, and Anne S Christin
- Subjects
Pharmacology ,0303 health sciences ,Adoptive cell transfer ,viruses ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Immunotherapy ,Biology ,Peripheral blood mononuclear cell ,Virology ,Virus ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Immunology ,Drug Discovery ,medicine ,Genetics ,Cytotoxic T cell ,Molecular Medicine ,Molecular Biology ,CD8 ,030304 developmental biology ,030215 immunology - Abstract
Severe and fatal viral infections remain common after hematopoietic stem cell transplantation. Adoptive transfer of cytotoxic T lymphocytes (CTLs) specific for Epstein–Barr virus (EBV), cytomegalovirus (CMV), and adenoviral antigens can treat infections that are impervious to conventional therapies, but broader implementation and extension to additional viruses is limited by competition between virus-derived antigens and time-consuming and laborious manufacturing procedures. We now describe a system that rapidly generates a single preparation of polyclonal (CD4+ and CD8+) CTLs that is consistently specific for 15 immunodominant and subdominant antigens derived from 7 viruses (EBV, CMV, Adenovirus (Adv), BK, human herpes virus (HHV)-6, respiratory syncytial virus (RSV), and Influenza) that commonly cause post-transplant morbidity and mortality. CTLs can be rapidly produced (10 days) by a single stimulation of donor peripheral blood mononuclear cells (PBMCs) with a peptide mixture spanning the target antigens in the presence of the potent prosurvival cytokines interleukin-4 (IL4) and IL7. This approach reduces the impact of antigenic competition with a consequent increase in the antigenic repertoire and frequency of virus-specific T cells. Our approach can be readily introduced into clinical practice and should be a cost-effective alternative to common antiviral prophylactic agents for allogeneic hematopoietic stem cell transplant (HSCT) recipients.
- Published
- 2012
- Full Text
- View/download PDF
44. A phase II study evaluating the safety and efficacy of an adenovirus-ΔLMP1-LMP2 transduced dendritic cell vaccine in patients with advanced metastatic nasopharyngeal carcinoma
- Author
-
W. K. Chia, W.-W. Wang, W. M. Tai, J. J. Chen, W. T. Lim, L. Sun, Stephen Gottschalk, Han Chong Toh, M. Teo, S. S. Leong, and E. H. Tan
- Subjects
Adult ,Male ,Epstein-Barr Virus Infections ,Genetic Vectors ,Nasopharyngeal neoplasm ,Kaplan-Meier Estimate ,medicine.disease_cause ,Cancer Vaccines ,Disease-Free Survival ,Adenoviridae ,Viral Matrix Proteins ,Immune system ,Antigen ,hemic and lymphatic diseases ,otorhinolaryngologic diseases ,medicine ,Humans ,Epstein–Barr virus infection ,Cells, Cultured ,Sequence Deletion ,Nasopharyngeal Carcinoma ,business.industry ,Carcinoma ,Nasopharyngeal Neoplasms ,Dendritic Cells ,Original Articles ,Hematology ,Dendritic cell ,Middle Aged ,medicine.disease ,Epstein–Barr virus ,Coculture Techniques ,stomatognathic diseases ,Treatment Outcome ,Oncology ,Nasopharyngeal carcinoma ,Delayed hypersensitivity ,Immunology ,Female ,business - Abstract
Background: Individuals with metastatic Epstein–Barr virus (EBV)-positive nasopharyngeal carcinoma (NPC) continue to have poor outcomes. To evaluate the ability of a dendritic cell (DC) vaccine to target subdominant EBV antigens LMP1 and LMP2 expressed by NPC cells, we vaccinated patients using autologous DCs transduced with an adenovirus encoding a truncated LMP1 (ΔLMP1) and full-length LMP2 (Ad-ΔLMP1-LMP2). Materials and methods: Sixteen subjects with metastatic NPC received Ad-ΔLMP1-LMP2 DC vaccines i.d. biweekly for up to five doses. Toxicity, immune responses and clinical responses were determined. Results: Most patients had extensive disease, with a median of three visceral sites of involvement (range 1–7). No significant toxicity was observed. Ad-ΔLMP1-LMP2 DCs induced delayed type hypersensitivity responses in 9 out of 12 patients, but although these DCs activated LMP1/2-specific T cells in vitro, no such increase in the frequency of peripheral LMP1/2-specific T cells was detected. Three patients had clinical responses including one with partial response (for 7½ months) and two with stable disease (for 6½ and 7½ months). Conclusions: Ad-ΔLMP1-LMP2 transduced DCs can be successfully generated and safely administered to patients with advanced NPC. Since efficacy was limited, future studies should focus on DC vaccines with greater potency administered to subjects with less tumor burden.
- Published
- 2012
45. The Use of Donor Lymphocyte Infusions As Prophylaxis and Treatment for Relapse in Children Post Hematopoietic Cell Transplant for Malignant Disease: A Single Institution Experience
- Author
-
Hao Liu, Tami John, Caridad Martinez, Bilal Omer, Nabil Ahmed, Carl E. Allen, Malcolm K. Brenner, Kathryn Leung, Jesse Wu, Stephen Gottschalk, Helen E. Heslop, Swati Naik, Meena Hegde, Ghadir Sasa, and Robert A. Krance
- Subjects
Oncology ,Transplantation ,medicine.medical_specialty ,Hematopoietic cell ,business.industry ,Lymphocyte ,Hematology ,Malignant disease ,medicine.anatomical_structure ,Internal medicine ,Immunology ,medicine ,Single institution ,business - Published
- 2017
46. Cytotoxic T Lymphocytes Simultaneously Targeting Multiple Tumor-associated Antigens to Treat EBV Negative Lymphoma
- Author
-
Anne S Christin, Alexandra Rousseau, Ulrike Gerdemann, Tamara Tripic, Malcolm K. Brenner, Catherine M. Bollard, Cliona M. Rooney, Usha L. Katari, Juan F. Vera, Ann M. Leen, Conrad Russell Y. Cruz, Helen E. Heslop, Stephen Gottschalk, and Barbara Savoldo
- Subjects
Adult ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Lymphoma ,medicine.medical_treatment ,Human leukocyte antigen ,Biology ,Lymphocyte Activation ,Epitope ,Monocytes ,Immunoenzyme Techniques ,03 medical and health sciences ,Epitopes ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,Peptide Library ,hemic and lymphatic diseases ,Drug Discovery ,medicine ,Genetics ,Cytotoxic T cell ,Humans ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,Pharmacology ,0303 health sciences ,PRAME ,Immunotherapy ,medicine.disease ,Flow Cytometry ,Coculture Techniques ,Peptide Fragments ,3. Good health ,030220 oncology & carcinogenesis ,Immunology ,Molecular Medicine ,Original Article ,CD8 ,T-Lymphocytes, Cytotoxic - Abstract
Although immunotherapy with Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) can treat EBV-associated Hodgkin and non-Hodgkin lymphoma (HL/NHL), more than 50% of such tumors are EBV negative. We now describe an approach that allows us to consistently generate, in a single line, CTLs that recognize a wide spectrum of nonviral tumor-associated antigens (TAAs) expressed by human HL/NHL, including Survivin, MAGE-A4, Synovial sarcoma X (SSX2), preferentially expressed antigen in melanoma (PRAME) and NY-ESO-1. We could generate these CTLs from nine of nine healthy donors and five of eight lymphoma patients, irrespective of human leukocyte antigen (HLA) type. We reactivated TAA-directed T cells ex vivo, by stimulation with dendritic cells (DCs) pulsed with overlapping peptide libraries spanning the chosen antigens in the presence of an optimized Th1-polarizing, prosurvival/proliferative and Treg inhibitory cytokine combination. The resultant lines of CD4(+) and CD8(+), polycytokine-producing T cells are directed against a multiplicity of epitopes expressed on the selected TAAs, with cytolytic activity against autologous tumor cells. Infusion of such multispecific monocultures may extend the benefits of CTL therapy to treatment even of EBV negative HL and NHL.
- Published
- 2011
- Full Text
- View/download PDF
47. PiggyBac-mediated Cancer Immunotherapy Using EBV-specific Cytotoxic T-cells Expressing HER2-specific Chimeric Antigen Receptor
- Author
-
Nabil Ahmed, Lisa Rollins, Gianpietro Dotti, Vita S. Salsman, Yozo Nakazawa, Leslie E. Huye, Cliona M. Rooney, Stephen Gottschalk, Ann M. Leen, and Matthew H. Wilson
- Subjects
Male ,Adoptive cell transfer ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Receptor, ErbB-2 ,medicine.medical_treatment ,T cell ,Recombinant Fusion Proteins ,Antigen presentation ,Antigens, CD19 ,Enzyme-Linked Immunosorbent Assay ,Mice, SCID ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Antigen ,Mice, Inbred NOD ,Transduction, Genetic ,Cell Line, Tumor ,Drug Discovery ,medicine ,Tumor Cells, Cultured ,Genetics ,Cytotoxic T cell ,Animals ,Humans ,skin and connective tissue diseases ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Antigen Presentation ,Brain Neoplasms ,Immunotherapy ,Flow Cytometry ,Molecular biology ,Chimeric antigen receptor ,Coculture Techniques ,3. Good health ,Receptors, Antigen ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,T-Lymphocytes, Cytotoxic - Abstract
Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes (CTLs) can be modified to function as heterologous tumor directed effector cells that survive longer in vivo than tumor directed T cells without virus specificity, due to chronic stimulation by viral antigens expressed during persistent infection in seropositive individuals. We evaluated the nonviral piggyBac (PB) transposon system as a platform for modifying EBV-CTLs to express a functional human epidermal growth factor receptor 2-specific chimeric antigen receptor (HER2-CAR) thereby directing virus-specific, gene modified CTLs towards HER2-positive cancer cells. Peripheral blood mononuclear cells (PBMCs) were nucleofected with transposons encoding a HER2-CAR and a truncated CD19 molecule for selection followed by specific activation and expansion of EBV-CTLs. HER2-CAR was expressed in ~40% of T cells after CD19 selection with retention of immunophenotype, polyclonality, and function. HER2-CAR-modified EBV-CTLs (HER2-CTLs) killed HER2-positive brain tumor cell lines in vitro, exhibited transient and reversible increases in HER2-CAR expression following antigen-specific stimulation, and stably expressed HER2-CAR beyond 120 days. Adoptive transfer of PB-modified HER2-CTLs resulted in tumor regression in a murine xenograft model. Our results demonstrate that PB can be used to redirect virus-specific CTLs to tumor targets, which should prolong tumor-specific T cell survival in vivo producing more efficacious immunotherapy.
- Published
- 2011
- Full Text
- View/download PDF
48. Expansion of T cells targeting multiple antigens of cytomegalovirus, Epstein–Barr virus and adenovirus to provide broad antiviral specificity after stem cell transplantation
- Author
-
Benjamin Tzou, Helen E. Heslop, C.M. Rooney, Hao Liu, Stephen Gottschalk, Patrick J. Hanley, Catherine M. Bollard, Conrad Russell Y. Cruz, Gail J. Demmler-Harrison, Stephanie Ku, and Donald R. Shaffer
- Subjects
Herpesvirus 4, Human ,Cancer Research ,Recombinant Fusion Proteins ,viruses ,medicine.medical_treatment ,Immunology ,Cell Culture Techniques ,Congenital cytomegalovirus infection ,Cytomegalovirus ,T-Cell Antigen Receptor Specificity ,Hematopoietic stem cell transplantation ,Biology ,Lymphocyte Activation ,medicine.disease_cause ,Article ,Virus ,Adenoviridae ,Immediate-Early Proteins ,Viral Matrix Proteins ,Postoperative Complications ,Antigen ,hemic and lymphatic diseases ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Cells, Cultured ,Genetics (clinical) ,Cell Proliferation ,Transplantation ,Hematopoietic Stem Cell Transplantation ,virus diseases ,Cell Biology ,Phosphoproteins ,medicine.disease ,Epstein–Barr virus ,Virology ,CTL ,surgical procedures, operative ,Oncology ,Virus Diseases ,Hematologic Neoplasms ,T-Lymphocytes, Cytotoxic - Abstract
Hematopoietic stem cell transplant (HSCT) is the treatment of choice for a proportion of patients with hematologic malignancies as well as for non-malignant diseases. However, viral infections, particularly Epstein-Barr virus (EBV), cytomegalovirus (CMV) and adenovirus (Ad), remain problematic after transplant despite the use of antiviral drugs. We have shown that cytotoxic T lymphocytes (CTL) generated against CMV-pp65, EBV and Ad antigens in a single culture are capable of controlling infections with all three viruses after HSCT. Although pp65-specific CTL have proved efficacious for the control of CMV infection, several reports highlight the importance of targeting additional CMV antigens.To expand multivirus-specific T cells with activity against both CMV-pp65 and CMV-IE-1, peripheral blood mononuclear cells (PBMC) were transduced with the adenoviral vector (Ad5f35-IE-1-I-pp65). After 9-12 days the CTL were restimulated with autologous EBV-transformed B cells transduced with the same Ad vector.After 18 days in culture nine CTL lines expanded from less than 1.5 × 10(7) PBMC to a mean of 6.1 × 10(7) T cells that recognized CMV antigens pp65 [median 273 spot-forming cells (SFC), range 47-995] and IE-1 (median 154 SFC, range 11-505), the Ad antigens hexon (median 153 SFC, range 26-465) and penton (median 37 SFC, range 1-353), as well as EBV lymphoblastoid cell lines (median 55 SFC, range 9-301). Importantly, the T cells recognized at least two antigens per virus and lysed virus peptide-pulsed targets.CTL that target at least two antigens each of CMV, EBV and Ad should have clinical benefit with broad coverage of all three viruses and enhanced control of CMV infections compared with current protocols.
- Published
- 2011
49. Good manufacturing practice-grade cytotoxic T lymphocytes specific for latent membrane proteins (LMP)-1 and LMP2 for patients with Epstein–Barr virus-associated lymphoma
- Author
-
Adrian P. Gee, Catherine M. Bollard, M. Helen Huls, Stephen Gottschalk, Cliona M. Rooney, and Ann M. Leen
- Subjects
Cancer Research ,Transplantation ,Immunology ,Cell Biology ,Biology ,medicine.disease ,medicine.disease_cause ,Epstein–Barr virus ,Virus ,Lymphoma ,CTL ,Oncology ,Membrane protein ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Antigen-presenting cell ,Epstein–Barr virus infection ,Genetics (clinical) - Abstract
In the last decade, virus specific CTL manufacture has become a more widespread and reproducible technology. A number of clinical grade techniques have been described. Here we outline the approach used at Baylor College of Medicine (BCM) since 2004 to treat over 35 patients with EBV-positive lymphoma using T cells specific for the latent membrane proteins (LMP1 and LMP2) of EBV in phase I/II clinical trials. Clinical outcomes have been described, but briefly objective tumor responses were achieved in 11/16 patients and complete remissions in 8. [1], [2]Here we outline the CTL manufacturing process. We have made the detailed SOPs required available with appropriate URL links throughout the manuscript. These should facilitate the creation of protocols suitable for regulatory approval and provide the basis for GMP manufacture of LMP1- and LMP2-specific T cells.
- Published
- 2011
50. Adverse events following infusion of T cells for adoptive immunotherapy: a 10-year experience
- Author
-
Cliona M. Rooney, Conrad Russell Y. Cruz, Patrick J. Hanley, Helen E. Heslop, Chrystal U. Louis, James A. Arce, Gianpietro Dotti, Yu Feng Lin, Malcolm K. Brenner, Barbara Savoldo, Hao Liu, Ann M. Leen, Catherine M. Bollard, Adrian P. Gee, Stephen Gottschalk, and Vicky Torrano
- Subjects
Cancer Research ,Fever ,Nausea ,T-Lymphocytes ,T cell ,Immunology ,Immunotherapy, Adoptive ,Article ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Infusions, Intravenous ,Adverse effect ,Home Infusion Therapy ,Genetics (clinical) ,Transplantation ,United States Food and Drug Administration ,business.industry ,Diphenhydramine ,Age Factors ,Drugs, Investigational ,Cell Biology ,Chills ,United States ,medicine.anatomical_structure ,Oncology ,Anesthesia ,Postoperative Nausea and Vomiting ,Vomiting ,Premedication ,medicine.symptom ,business ,Follow-Up Studies ,medicine.drug - Abstract
Background aims The Food and Drug Administration (FDA) currently recommends at least 4h of recipient monitoring after T cell infusions to detect early infusion reactions. Recent catastrophic reactions to ‘first-in-man' biologic agents have emphasized the importance of this rule for initial studies of new products. The value of such monitoring for better established agents is less obvious. Methods We reviewed infusion-related adverse events (AE) following administration of ex vivo -expanded T cell products (antigen-specific cytotoxic T lymphocytes, allodepleted T cells, and genetically modified T cells) on investigational new drug (IND) studies in our center. Results From 1998 to 2008, we infused 381 T cell products to 180 recipients, enrolled on 18 studies, receiving T cells targeting malignancies or post-transplant viral infections. There were no grade 3–4 infusion reactions during initial monitoring or 24-h follow-up. Twenty-four mild (grade 1–2) AE occurred in 21 infusions either during or immediately following infusion (up to 6h), most commonly nausea and vomiting (10/24, 41.6%), probably because of the dimethyl sulfoxide cryoprotectant, and hypotension (20.8%), attributable to diphenhydramine pre-medication. Twenty-two additional non-severe events were reported within 24h of infusion, most commonly culture-negative fever, chills and nausea. An increased risk of adverse events was associated with age [incidence rate ratio (IRR) 0.98; 95% confidence interval (CI) 0.96–1.00, P =0.05], while an increased risk of immediate infusion-related events was higher in patients reporting allergies (IRR 2.72, 95% CI 1.00–7.40, P =0.05); sex, disease type and T cell source (allogeneic or autologous) had no effect on frequency of adverse events. Conclusions Infusion of these T cell products was safe in the outpatient setting and associated with no severe reactions, so monitoring for 1h after infusion is probably sufficient. As many of the AE were attributable to diphenhydramine premedication, a lower dose (0.25mg/kg) should be selected.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.