Lymphodepletion – an essential but undervalued part of the chimeric antigen receptor T-cell therapy cycle

Lymphodepletion (LD) or conditioning is an essential step in the application of currently used autologous and allogeneic chimeric antigen receptor T-cell (CAR-T) therapies as it maximizes engraftment, efficacy and long-term survival of CAR-T. Its main modes of action are the depletion and modulation of endogenous lymphocytes, conditioning of the microenvironment for improved CAR-T expansion and persistence, and reduction of tumor load. However, most LD regimens provide a broad and fairly unspecific suppression of T-cells as well as other hematopoietic cells, which can also lead to severe side effects, particularly infections. We reviewed 1271 published studies (2011-2023) with regard to current LD strategies for approved anti-CD19 CAR-T products for large B cell lymphoma (LBCL). Fludarabine (Flu) and cyclophosphamide (Cy) (alone or in combination) were the most commonly used agents. A large number of different schemes and combinations have been reported. In the respective schemes, doses of Flu and Cy (range 75-120mg/m2 and 750-1.500mg/m2) and wash out times (range 2-5 days) differed substantially. Furthermore, combinations with other agents such as bendamustine (benda), busulfan or alemtuzumab (for allogeneic CAR-T) were described. This diversity creates a challenge but also an opportunity to investigate the impact of LD on cellular kinetics and clinical outcomes of CAR-T. Only 21 studies explicitly investigated in more detail the influence of LD on safety and efficacy. As Flu and Cy can potentially impact both the in vivo activity and toxicity of CAR-T, a more detailed analysis of LD outcomes will be needed before we are able to fully assess its impact on different T-cell subsets within the CAR-T product. The T2EVOLVE consortium propagates a strategic investigation of LD protocols for the development of optimized conditioning regimens.

[1]  B. Sahaf,et al.  Bendamustine vs. fludarabine/cyclophosphamide lymphodepletion prior to BCMA CAR-T cell therapy in multiple myeloma , 2023, Blood cancer journal.

[2]  N. Ajami,et al.  Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring. , 2023, Cancer cell.

[3]  S. Ong,et al.  Bendamustine lymphodepletion is a well‐tolerated alternative to fludarabine and cyclophosphamide lymphodepletion for axicabtagene ciloleucel therapy for aggressive B‐cell lymphoma , 2023, American journal of hematology.

[4]  E. Giné,et al.  P1394: RITUXIMAB BASED LYMPHODEPLETION MAY INCREASE ENGRAFTMENT AND EFFICACY OF SECOND INFUSIONS OF CART19 CELLS IN B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA. , 2023, HemaSphere.

[5]  M. Juan,et al.  Fractionated initial infusion and booster dose of ARI0002h, a humanised, BCMA-directed CAR T-cell therapy, for patients with relapsed or refractory multiple myeloma (CARTBCMA-HCB-01): a single-arm, multicentre, academic pilot study. , 2023, The Lancet. Oncology.

[6]  R. Houot,et al.  Immune Effector Cell-Associated Hematotoxicity (ICAHT): EHA/EBMT Consensus Grading and Best Practice Recommendations. , 2023, Blood.

[7]  J. Kochenderfer,et al.  Long-term outcomes following CAR T cell therapy: what we know so far , 2023, Nature Reviews Clinical Oncology.

[8]  Stacy L. Gelhaus,et al.  Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment , 2023, Cell.

[9]  N. Hijiya,et al.  Clofarabine for Lymphodepletion Before CAR-T-Cell Infusion: A Brief Case Report , 2023, Journal of pediatric hematology/oncology.

[10]  B. Baradaran,et al.  The current landscape of CAR T-cell therapy for solid tumors: Mechanisms, research progress, challenges, and counterstrategies , 2023, Frontiers in Immunology.

[11]  E. Elinav,et al.  A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy , 2023, Nature Medicine.

[12]  Alessandra M. Sullivan,et al.  Preclinical proof of concept for VivoVec, a lentiviral-based platform for in vivo CAR T-cell engineering , 2023, Journal for ImmunoTherapy of Cancer.

[13]  M. Ayas,et al.  Clofarabine in Pediatric Acute Relapsed or Refractory Leukemia: Where Do We Stand on the Bridge to Hematopoietic Stem Cell Transplantation? , 2023, Journal of hematology.

[14]  M. Liedtke,et al.  Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results , 2023, Nature Medicine.

[15]  P. Riedell,et al.  Population Pharmacokinetic Model Identifies an Optimal Fludarabine Exposure for Improved Outcomes after CD19-Directed CAR T Cell Therapy for Aggressive B-NHL: Analysis from the Cell Therapy Consortium , 2022, Blood.

[16]  Y. Taur,et al.  Author Correction: Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy , 2022, Nature Medicine.

[17]  Matthew J. Frigault,et al.  UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukaemia (CALM): a phase 1, dose-escalation trial. , 2022, The Lancet. Haematology.

[18]  H. Mollenkopf,et al.  Selected commensals educate the intestinal vascular and immune system for immunocompetence , 2022, Microbiome.

[19]  R. Houot,et al.  A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma , 2022, Nature Medicine.

[20]  F. Locke,et al.  Outcomes of CD19 targeted CAR T-cell Therapy for Patients with Reduced Renal Function Including Dialysis. , 2022, Transplantation and cellular therapy.

[21]  U. Jäger,et al.  Bendamustine is Safe and Effective for Lymphodepletion Before Tisagenlecleucel in Patients with Refractory or Relapsed Large B-Cell Lymphomas. , 2022, Annals of oncology : official journal of the European Society for Medical Oncology.

[22]  J. Byrd,et al.  Anti-CD19 CAR T cells in combination with ibrutinib for the treatment of chronic lymphocytic leukemia , 2022, Blood advances.

[23]  C. Ramos,et al.  Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion , 2022, Nature Reviews Clinical Oncology.

[24]  B. Baradaran,et al.  A Systematic Review on PD-1 Blockade and PD-1 Gene-Editing of CAR-T Cells for Glioma Therapy: From Deciphering to Personalized Medicine , 2022, Frontiers in Immunology.

[25]  D. Weissman,et al.  CAR T cells produced in vivo to treat cardiac injury , 2022, Science.

[26]  R. de Waal Malefyt,et al.  An orthogonal IL-2 and IL-2Rβ system drives persistence and activation of CAR T cells and clearance of bulky lymphoma , 2021, Science Translational Medicine.

[27]  M. Kersten,et al.  Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial , 2021, Nature Medicine.

[28]  G. Salles,et al.  Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. , 2021, The Lancet. Oncology.

[29]  Snehit Prabhu,et al.  Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy , 2021, Blood advances.

[30]  A. Huitema,et al.  Fludarabine exposure predicts outcome after CD19 CAR T-cell therapy in children and young adults with acute leukemia , 2021, Blood advances.

[31]  U. Jaeger,et al.  Bendamustine Is a Safe and Effective Regimen for Lymphodepletion before Tisagenlecleucel in Patients with Large B-Cell Lymphomas , 2021, Blood.

[32]  P. Riedell,et al.  Phase 1 TRANSCEND CLL 004 study of lisocabtagene maraleucel in patients with relapsed/refractory CLL or SLL , 2021, Blood.

[33]  M. Hallek,et al.  Chronic lymphocytic leukemia: 2022 update on diagnostic and therapeutic procedures , 2021, American journal of hematology.

[34]  M. Abbasian,et al.  Durable Responses and Low Toxicity After Fast Off-Rate CD19 Chimeric Antigen Receptor-T Therapy in Adults With Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[35]  M. Perales,et al.  Modified-EASIX predicts severe cytokine release syndrome and neurotoxicity after Chimeric Antigen Receptor (CAR) T cells. , 2021, Blood advances.

[36]  M. Kersten,et al.  Efficacy and safety of CD19‐directed CAR‐T cell therapies in patients with relapsed/refractory aggressive B‐cell lymphomas: Observations from the JULIET, ZUMA‐1, and TRANSCEND trials , 2021, American journal of hematology.

[37]  T. Gress,et al.  Microbial short-chain fatty acids modulate CD8+ T cell responses and improve adoptive immunotherapy for cancer , 2021, Nature Communications.

[38]  E. Hoster,et al.  CAR-HEMATOTOX: A model for CAR T-cell related hematological toxicity in relapsed/refractory large B-cell lymphoma. , 2021, Blood.

[39]  S. Jagannath,et al.  Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study , 2021, The Lancet.

[40]  R. Houot,et al.  KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study , 2021, The Lancet.

[41]  R. Simpson,et al.  Immunomodulatory Effects of Bendamustine in Hematopoietic Cell Transplantation , 2021, Cancers.

[42]  S. Steinberg,et al.  Long-Term Follow-Up of CD19-CAR T-Cell Therapy in Children and Young Adults With B-ALL , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[43]  H. Goldschmidt,et al.  Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. , 2021, The New England journal of medicine.

[44]  J. Maher,et al.  Crosstown Traffic: Lymphodepleting Chemotherapy Drives CAR T Cells. , 2020, Cancer cell.

[45]  A. M. Houghton,et al.  Immunogenic Chemotherapy Enhances Recruitment of CAR-T Cells to Lung Tumors and Improves Antitumor Efficacy when Combined with Checkpoint Blockade. , 2020, Cancer cell.

[46]  E. Giné,et al.  CART19-BE-01: A Multicenter Trial of ARI-0001 Cell Therapy in Patients with CD19+ Relapsed/Refractory Malignancies. , 2020, Molecular therapy : the journal of the American Society of Gene Therapy.

[47]  Michael L. Wang,et al.  Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study , 2020, The Lancet.

[48]  J. Serody,et al.  Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[49]  H. Abken,et al.  TRUCKS, the fourth‐generation CAR T cells: Current developments and clinical translation , 2020, ADVANCES IN CELL AND GENE THERAPY.

[50]  Michael L. Wang,et al.  KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. , 2020, The New England journal of medicine.

[51]  A. Huitema,et al.  A semi‐mechanistic model based on glutathione depletion to describe intra‐individual reduction in busulfan clearance , 2020, British journal of clinical pharmacology.

[52]  Oliver J. Klein,et al.  An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors , 2020, Science.

[53]  David A. Williams,et al.  Toxicity and Response following CD19-specific CAR T cells in pediatric/young adult relapsed/refractory B-ALL. , 2019, Blood.

[54]  S. Grupp,et al.  Optimizing Chimeric Antigen Receptor T-Cell Therapy for Adults With Acute Lymphoblastic Leukemia. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[55]  A. d’Arminio Monforte,et al.  Disseminated cytomegalovirus disease after bendamustine: a case report and analysis of circulating B- and T-cell subsets , 2019, BMC Infectious Diseases.

[56]  A. Huitema,et al.  Fludarabine exposure in the conditioning prior to allogeneic hematopoietic cell transplantation predicts outcomes. , 2019, Blood advances.

[57]  Y. Shimizu Faculty Opinions recommendation of An effective mouse model for adoptive cancer immunotherapy targeting neoantigens. , 2019, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[58]  N. Restifo,et al.  An effective mouse model for adoptive cancer immunotherapy targeting neoantigens. , 2019, JCI insight.

[59]  D. Maloney,et al.  The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. , 2019, Blood.

[60]  S. Neelapu CAR-T efficacy: is conditioning the key? , 2019, Blood.

[61]  D. Maloney,et al.  Factors associated with durable EFS in adult B-cell ALL patients achieving MRD-negative CR after CD19 CAR T-cell therapy. , 2019, Blood.

[62]  Boris Lamp,et al.  The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes , 2019, Nature Communications.

[63]  G. Salles,et al.  Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B‐Cell Lymphoma , 2019, The New England journal of medicine.

[64]  I. Flinn,et al.  Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. , 2019, The Lancet. Oncology.

[65]  M. Sadelain,et al.  Low-Dose Radiation Conditioning Enables CAR T Cells to Mitigate Antigen Escape. , 2018, Molecular therapy : the journal of the American Society of Gene Therapy.

[66]  A. Huitema,et al.  Population Pharmacokinetics of Fludarabine in Children and Adults during Conditioning Prior to Allogeneic Hematopoietic Cell Transplantation , 2018, Clinical Pharmacokinetics.

[67]  S. Mehrotra,et al.  Enhanced Lymphodepletion Is Insufficient to Replace Exogenous IL2 or IL15 Therapy in Augmenting the Efficacy of Adoptively Transferred Effector CD8+ T Cells. , 2018, Cancer research.

[68]  M. Spitzer,et al.  Engineered Tumor-Targeted T Cells Mediate Enhanced Anti-Tumor Efficacy Both Directly and through Activation of the Endogenous Immune System , 2018, Cell reports.

[69]  Hans Bitter,et al.  Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia , 2018, Nature Medicine.

[70]  C. June,et al.  CAR T cell immunotherapy for human cancer , 2018, Science.

[71]  K. Tamada,et al.  IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor , 2018, Nature Biotechnology.

[72]  K. Davis,et al.  Tisagenlecleucel in Children and Young Adults with B‐Cell Lymphoblastic Leukemia , 2018, The New England journal of medicine.

[73]  Mithat Gonen,et al.  Long‐Term Follow‐up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia , 2018, The New England journal of medicine.

[74]  D. Gilham,et al.  CD19 CAR T Cells Expressing IL-12 Eradicate Lymphoma in Fully Lymphoreplete Mice through Induction of Host Immunity , 2017, Molecular therapy oncolytics.

[75]  R. Levy,et al.  Axicabtagene Ciloleucel CAR T‐Cell Therapy in Refractory Large B‐Cell Lymphoma , 2017, The New England journal of medicine.

[76]  Daniel Li,et al.  Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. , 2017, Blood.

[77]  R. Kruse,et al.  Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells. , 2017, Cancer discovery.

[78]  J. Byrd,et al.  Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated With CD19-Specific Chimeric Antigen Receptor-Modified T Cells After Failure of Ibrutinib. , 2017, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[79]  S. Riddell,et al.  Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. , 2017, Blood.

[80]  Michel Sadelain,et al.  Therapeutic T cell engineering , 2017, Nature.

[81]  C. van Kesteren,et al.  Association between anti-thymocyte globulin exposure and survival outcomes in adult unrelated haemopoietic cell transplantation: a multicentre, retrospective, pharmacodynamic cohort analysis. , 2017, The Lancet. Haematology.

[82]  S. Heimfeld,et al.  Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor–modified T cells , 2016, Science Translational Medicine.

[83]  M. Sabatino,et al.  Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. , 2016, Blood.

[84]  Daniel Li,et al.  CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. , 2016, The Journal of clinical investigation.

[85]  E. Kimby,et al.  Optimal use of bendamustine in hematologic disorders: Treatment recommendations from an international consensus panel – an update , 2015, Leukemia & lymphoma.

[86]  H. Döhner,et al.  Outcome of advanced chronic lymphocytic leukemia following different first-line and relapse therapies: a meta-analysis of five prospective trials by the German CLL Study Group (GCLLSG) , 2015, Haematologica.

[87]  D. Maloney,et al.  Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo , 2015, Leukemia.

[88]  David L. Porter,et al.  Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia , 2015, Science Translational Medicine.

[89]  L. Specht,et al.  Modern radiation therapy for extranodal lymphomas: field and dose guidelines from the International Lymphoma Radiation Oncology Group. , 2015, International journal of radiation oncology, biology, physics.

[90]  S. Steinberg,et al.  T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial , 2015, The Lancet.

[91]  Pamela A Shaw,et al.  Chimeric antigen receptor T cells for sustained remissions in leukemia. , 2014, The New England journal of medicine.

[92]  B. Sandmaier,et al.  Conditioning regimens for hematopoietic cell transplantation: one size does not fit all. , 2014, Blood.

[93]  H. Eich,et al.  Modern radiation therapy for Hodgkin lymphoma: field and dose guidelines from the international lymphoma radiation oncology group (ILROG). , 2014, International journal of radiation oncology, biology, physics.

[94]  J. Burke,et al.  Randomized trial of bendamustine-rituximab or R-CHOP/R-CVP in first-line treatment of indolent NHL or MCL: the BRIGHT study. , 2014, Blood.

[95]  T. Cooper,et al.  Whole-Body Irradiation Increases the Magnitude and Persistence of Adoptively Transferred T Cells Associated with Tumor Regression in a Mouse Model of Prostate Cancer , 2014, Cancer Immunology Research.

[96]  A. Ng,et al.  Modern radiation therapy for nodal non-Hodgkin lymphoma-target definition and dose guidelines from the International Lymphoma Radiation Oncology Group. , 2014, International journal of radiation oncology, biology, physics.

[97]  Qing He,et al.  Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia , 2014, Science Translational Medicine.

[98]  Michel Sadelain,et al.  CD19 CAR-Targeted T Cells Induce Long-Term Remission and B Cell Aplasia in an Immunocompetent Mouse Model of B Cell Acute Lymphoblastic Leukemia , 2013, PloS one.

[99]  C. Guillemette,et al.  Overexpression of uridine diphospho glucuronosyltransferase 2B17 in high-risk chronic lymphocytic leukemia. , 2013, Blood.

[100]  Jinjuan Wang,et al.  CD20-specific adoptive immunotherapy for lymphoma using a chimeric antigen receptor with both CD28 and 4-1BB domains: pilot clinical trial results. , 2012, Blood.

[101]  Michel Sadelain,et al.  Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. , 2011, Blood.

[102]  H. Döhner,et al.  Bendamustine combined with rituximab in patients with relapsed and/or refractory chronic lymphocytic leukemia: a multicenter phase II trial of the German Chronic Lymphocytic Leukemia Study Group. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[103]  A. Bagg,et al.  Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. , 2011, The New England journal of medicine.

[104]  David L. Porter,et al.  T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia , 2011, Science Translational Medicine.

[105]  W. Wilson,et al.  Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. , 2010, Blood.

[106]  S. Rosenberg,et al.  Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. , 2010, Blood.

[107]  R. Greil,et al.  Fludarabine modulates composition and function of the T cell pool in patients with chronic lymphocytic leukaemia , 2010, Cancer Immunology, Immunotherapy.

[108]  M. Kalos,et al.  Antitransgene rejection responses contribute to attenuated persistence of adoptively transferred CD20/CD19-specific chimeric antigen receptor redirected T cells in humans. , 2010, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[109]  M. Sadelain,et al.  Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[110]  M. Atkins,et al.  Cytokine working group study of lymphodepleting chemotherapy, interleukin-2, and granulocyte-macrophage colony-stimulating factor in patients with metastatic melanoma: clinical outcomes and peripheral-blood cell recovery. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[111]  S. Dovedi,et al.  Natural Expression of the CD19 Antigen Impacts the Long-Term Engraftment but Not Antitumor Activity of CD19-Specific Engineered T Cells , 2010, The Journal of Immunology.

[112]  T. Pavlík,et al.  Efficacious but insidious: a retrospective analysis of fludarabine-induced myelotoxicity using long-term culture-initiating cells in 100 follicular lymphoma patients. , 2009, Experimental hematology.

[113]  T. Tani,et al.  Pre-treatment with cyclophosphamide or OX40 (CD134) costimulation targeting regulatory T cell function enhances the anti-tumor immune effect of adoptively transferred CD8+ T cells from wild-type mice. , 2009, Molecular medicine reports.

[114]  C. Yee,et al.  Fludarabine Modulates Immune Response and Extends In Vivo Survival of Adoptively Transferred CD8 T Cells in Patients with Metastatic Melanoma , 2009, PloS one.

[115]  Brian R. Phillips,et al.  A novel phenotypic method to determine fludarabine triphosphate accumulation in T-lymphocytes from hematopoietic cell transplantation patients , 2009, Cancer Chemotherapy and Pharmacology.

[116]  S. Rosenberg,et al.  Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[117]  Jinjuan Wang,et al.  Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. , 2008, Blood.

[118]  J. Mulé,et al.  Nonmyeloablative Chemotherapy Followed by T-cell Adoptive Transfer and Dendritic Cell-based Vaccination Results in Rejection of Established Melanoma , 2008, Journal of immunotherapy.

[119]  Christian Stemberger,et al.  A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets. , 2007, Immunity.

[120]  V. Maguer-Satta,et al.  In Vitro Toxicity of Fludarabine on Normal Bone Marrow (BM) Progenitors Suggests Its Involvement in the Impairment of Hematopoietic Progenitor (HP) Mobilization. , 2007 .

[121]  S. Rosenberg,et al.  Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. , 2007, The Journal of clinical investigation.

[122]  S. Rosenberg,et al.  Hematopoietic stem cells promote the expansion and function of adoptively transferred antitumor CD8 T cells. , 2007, The Journal of clinical investigation.

[123]  J. Delabie,et al.  Standard CHOP-21 as first line therapy for elderly patients with Hodgkin's lymphoma , 2007, Leukemia & lymphoma.

[124]  S. Rosenberg,et al.  Increased intensity lymphodepletion and adoptive immunotherapy—how far can we go? , 2006, Nature Clinical Practice Oncology.

[125]  C. Klebanoff,et al.  Interleukin-2-Dependent Mechanisms of Tolerance and Immunity In Vivo1 , 2006, The Journal of Immunology.

[126]  A. Zelenetz,et al.  Pentostatin, cyclophosphamide, and rituximab is an active, well-tolerated regimen for patients with previously treated chronic lymphocytic leukemia. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[127]  B. D. de Pauw,et al.  Microbiologic consequences of new approaches to managing hematologic malignancies. , 2005, Reviews in clinical and experimental hematology.

[128]  N. Schmitz,et al.  Frontline therapy with rituximab added to the combination of cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) significantly improves the outcome for patients with advanced-stage follicular lymphoma compared with therapy with CHOP alone: results of a prospective randomized study of t , 2005, Blood.

[129]  G. Plautz,et al.  Host lymphodepletion augments T cell adoptive immunotherapy through enhanced intratumoral proliferation of effector cells. , 2005, Cancer research.

[130]  S. Rosenberg,et al.  Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells , 2005, The Journal of experimental medicine.

[131]  Xiaojing Ma,et al.  Interleukin-12: an update on its immunological activities, signaling and regulation of gene expression. , 2005, Current immunology reviews.

[132]  N. Restifo,et al.  Less is more: lymphodepletion followed by hematopoietic stem cell transplant augments adoptive T-cell-based anti-tumor immunotherapy. , 2005, Current opinion in immunology.

[133]  J. Schlom,et al.  Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. , 2005, Blood.

[134]  S. Rosenberg,et al.  Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[135]  S. Rosenberg,et al.  CD8+ T Cell Immunity Against a Tumor/Self-Antigen Is Augmented by CD4+ T Helper Cells and Hindered by Naturally Occurring T Regulatory Cells , 2005, The Journal of Immunology.

[136]  C. Klebanoff,et al.  Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. , 2005, Trends in immunology.

[137]  George Coukos,et al.  Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival , 2004, Nature Medicine.

[138]  B. Chauffert,et al.  CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative , 2004, European journal of immunology.

[139]  M. Raffeld,et al.  Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes , 2002, Science.

[140]  C. June,et al.  Cutting Edge: Regulatory T Cells from Lung Cancer Patients Directly Inhibit Autologous T Cell Proliferation1 , 2002, The Journal of Immunology.

[141]  N. Hayashi,et al.  Administration of interleukin-12 enhances the therapeutic efficacy of dendritic cell-based tumor vaccines in mouse hepatocellular carcinoma. , 2001, Cancer research.

[142]  Jianzhu Chen,et al.  Homeostasis-Stimulated Proliferation Drives Naive T Cells to Differentiate Directly into Memory T Cells , 2000, The Journal of experimental medicine.

[143]  L. Bogatzki,et al.  Naive T Cells Transiently Acquire a Memory-like Phenotype during Homeostasis-Driven Proliferation , 2000, The Journal of experimental medicine.

[144]  R. Ahmed,et al.  Cutting Edge: Naive T Cells Masquerading as Memory Cells , 2000, The Journal of Immunology.

[145]  J. Shimizu,et al.  Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. , 1999, Journal of immunology.

[146]  J. Sprent,et al.  The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. , 1999, Immunity.

[147]  K. Brduscha-Riem,et al.  Naïve cytotoxic T lymphocytes spontaneously acquire effector function in lymphocytopenic recipients: A pitfall for T cell memory studies? , 1999, European journal of immunology.

[148]  E. Proietti,et al.  Importance of cyclophosphamide-induced bystander effect on T cells for a successful tumor eradication in response to adoptive immunotherapy in mice. , 1998, The Journal of clinical investigation.

[149]  Julie C. Adkins,et al.  Fludarabine. An update of its pharmacology and use in the treatment of haematological malignancies. , 1997, Drugs.

[150]  B. Rocha,et al.  Peripheral selection of the T cell repertoire. , 1991, Science.

[151]  Z. Eshhar,et al.  Chimeric T cell receptor which incorporates the anti-tumour specificity of a monoclonal antibody with the cytolytic activity of T cells: a model system for immunotherapeutical approach. , 1990, The British journal of cancer. Supplement.

[152]  Z. Eshhar,et al.  Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[153]  M. Drayson,et al.  The stable and permanent expansion of functional T lymphocytes in athymic nude rats after a single injection of mature T cells. , 1987, Journal of immunology.

[154]  S. Rosenberg,et al.  A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. , 1986, Science.

[155]  R. North Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on elimination of tumor-induced suppressor T cells , 1982, The Journal of experimental medicine.

[156]  P. Greenberg,et al.  Specificity of adoptive chemoimmunotherapy of established syngeneic tumors. , 1980, Journal of immunology.

[157]  A. Einstein,et al.  Syngeneic adoptive immunotherapy and chemoimmunotherapy of a Friend leukemia: requirement for T cells. , 1975, Journal of immunology.

[158]  M. Perales,et al.  Modi fi ed EASIX predicts severe cytokine release syndrome and neurotoxicity after chimeric antigen receptor T cells , 2021 .

[159]  J. Markmann,et al.  Homeostatic proliferation is a barrier to transplantation tolerance , 2004, Nature Medicine.

[160]  M. Hallek,et al.  Chemotherapy combination treatment regimens with fludarabine in chronic lymphocytic leukemia. , 2004, The hematology journal : the official journal of the European Haematology Association.

[161]  A. Sparreboom,et al.  Repeated administrations of interleukin (IL)-12 are associated with persistently elevated plasma levels of IL-10 and declining IFN-gamma, tumor necrosis factor-alpha, IL-6, and IL-8 responses. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[162]  Pierre Morel,et al.  CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. , 2002, The New England journal of medicine.