Hematopoietic stem cells promote the expansion and function of adoptively transferred antitumor CD8 T cells.

Depleting host immune elements with nonmyeloablative regimens prior to the adoptive transfer of tumor-specific CD8(+) T cells significantly enhances tumor treatment. In the current study, superior antitumor efficacy was achieved by further increasing the intensity of lymphodepletion to a level that required HSC transplantation. Surprisingly, the HSC transplant and not the increased lymphodepletion caused a robust expansion of adoptively transferred tumor-specific CD8(+) T cells. The HSC-driven cell expansion of effector, but not of naive, CD8(+) T cells was independent of in vivo restimulation by MHC class I-expressing APCs. Simultaneously, HSCs also facilitated the reconstitution of the host lymphoid compartment, including inhibitory elements, not merely via the production of progeny cells but by enhancing the expansion of cells that had survived lymphodepletion. Profound lymphodepletion, by myeloablation or by genetic means, focused the nonspecific HSC boost preferentially toward the transferred tumor-specific T cells, leading to successful tumor treatment. These findings indicate that CD8(+) T cell-mediated tumor responses can be efficiently driven by HSCs in the myeloablative setting and have substantial implications for the design of new antitumor immunotherapies.

[1]  C. Rooney,et al.  Improving T cell therapy for cancer , 2006, Annual review of immunology.

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

[3]  S. Rosenberg,et al.  Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes , 2006, Science.

[4]  A. Frey Myeloid suppressor cells regulate the adaptive immune response to cancer. , 2006, The Journal of clinical investigation.

[5]  Paolo Serafini,et al.  Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. , 2006, The Journal of clinical investigation.

[6]  S. Quezada,et al.  CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. , 2006, The Journal of clinical investigation.

[7]  C. Klebanoff,et al.  CD8+ T‐cell memory in tumor immunology and immunotherapy , 2006, Immunological reviews.

[8]  K. Camphausen,et al.  Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy , 2006, The Journal of experimental medicine.

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

[10]  S. Rosenberg,et al.  Adoptive immunotherapy for cancer: building on success , 2006, Nature Reviews Immunology.

[11]  Gerd Ritter,et al.  Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[12]  M. A. Curotto de Lafaille,et al.  Control of homeostatic proliferation by regulatory T cells. , 2005, The Journal of clinical investigation.

[13]  W. Blackwelder,et al.  Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer , 2005, Nature Medicine.

[14]  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.

[15]  T. Fry,et al.  The Many Faces of IL-7: From Lymphopoiesis to Peripheral T Cell Maintenance , 2005, The Journal of Immunology.

[16]  S. Rosenberg,et al.  Acquisition of full effector function in vitro paradoxically impairs the in vivo antitumor efficacy of adoptively transferred CD8+ T cells. , 2005, The Journal of clinical investigation.

[17]  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.

[18]  W. Anderson,et al.  Soluble factor(s) from bone marrow cells can rescue lethally irradiated mice by protecting endogenous hematopoietic stem cells. , 2005, Experimental hematology.

[19]  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.

[20]  C. Klebanoff,et al.  Vaccine-Stimulated, Adoptively Transferred CD8+ T Cells Traffic Indiscriminately and Ubiquitously while Mediating Specific Tumor Destruction1 , 2004, The Journal of Immunology.

[21]  R. Boyd,et al.  Strategies to enhance T-cell reconstitution in immunocompromised patients , 2004, Nature Reviews Immunology.

[22]  J. Schlom,et al.  Sublethal Irradiation of Human Tumor Cells Modulates Phenotype Resulting in Enhanced Killing by Cytotoxic T Lymphocytes , 2004, Cancer Research.

[23]  A. Houghton,et al.  Concomitant Tumor Immunity to a Poorly Immunogenic Melanoma Is Prevented by Regulatory T Cells , 2004, The Journal of experimental medicine.

[24]  S. Rosenberg,et al.  Cancer immunotherapy: moving beyond current vaccines , 2004, Nature Medicine.

[25]  T. Waldmann,et al.  IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T Cells , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[26]  Michel Drouet,et al.  Single administration of stem cell factor, FLT-3 ligand, megakaryocyte growth and development factor, and interleukin-3 in combination soon after irradiation prevents nonhuman primates from myelosuppression: long-term follow-up of hematopoiesis. , 2004, Blood.

[27]  B. Barlogie,et al.  Treatment of multiple myeloma. , 2004, Blood.

[28]  Irving L Weissman,et al.  Biology of hematopoietic stem cells and progenitors: implications for clinical application. , 2003, Annual review of immunology.

[29]  R. V. van Lier,et al.  IL-15 induces antigen-independent expansion and differentiation of human naive CD8+ T cells in vitro. , 2003, Blood.

[30]  S. Rosenberg,et al.  Tumor Regression and Autoimmunity after Reversal of a Functionally Tolerant State of Self-reactive CD8+ T Cells , 2003, The Journal of experimental medicine.

[31]  M. Farrar,et al.  In Vivo Survival and Homeostatic Proliferation of Natural Killer Cells , 2003, The Journal of experimental medicine.

[32]  P. Burkett,et al.  Interleukin (IL)-15Rα–deficient Natural Killer Cells Survive in Normal but Not IL-15Rα–deficient Mice , 2003, The Journal of experimental medicine.

[33]  Leo Lefrançois,et al.  Cytokine control of memory T-cell development and survival , 2003, Nature Reviews Immunology.

[34]  P. Bourin,et al.  Short-term injection of antiapoptotic cytokine combinations soon after lethal gamma -irradiation promotes survival. , 2003, Blood.

[35]  George Coukos,et al.  Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. , 2003, The New England journal of medicine.

[36]  J. Thompson,et al.  Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: In vivo persistence, migration, and antitumor effect of transferred T cells , 2002, Proceedings of the National Academy of Sciences of the United States of America.

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

[38]  A. Swerdlow,et al.  Treatment of Epstein-Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells , 2002, The Lancet.

[39]  J. Mulé,et al.  Making room for T cells. , 2002, The Journal of clinical investigation.

[40]  C. Benoist,et al.  Cytokine Requirements for Acute and Basal Homeostatic Proliferation of Naive and Memory CD8+ T Cells , 2002, The Journal of experimental medicine.

[41]  J. Sprent,et al.  Interleukin (IL)-15 and IL-7 Jointly Regulate Homeostatic Proliferation of Memory Phenotype CD8+ Cells but Are Not Required for Memory Phenotype CD4+ Cells , 2002, The Journal of experimental medicine.

[42]  W. Murphy,et al.  The role of growth hormone in T‐cell development and reconstitution , 2002, Journal of leukocyte biology.

[43]  M. Tokuda,et al.  Growth hormone restores glucocorticoid‐induced T cell suppression , 2001, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[44]  S. Ansell,et al.  Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. , 2001, Blood.

[45]  Richard Murray,et al.  IL-7 is critical for homeostatic proliferation and survival of naïve T cells , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[46]  S. Jameson,et al.  Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo , 2000, Nature Immunology.

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

[48]  S. Jameson,et al.  Homeostatic expansion and phenotypic conversion of naïve T cells in response to self peptide/MHC ligands. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[49]  M. Bevan,et al.  Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts. , 1999, Immunity.

[50]  C. Janeway,et al.  Designing and maintaining the mature TCR repertoire: the continuum of self-peptide:self-MHC complex recognition. , 1999, Immunity.

[51]  S. Rosenberg,et al.  Apoptotic death of CD8+ T lymphocytes after immunization: induction of a suppressive population of Mac-1+/Gr-1+ cells. , 1998, Journal of immunology.

[52]  A. Burroughs,et al.  Reconstitution of EBV-specific T cell immunity in solid organ transplant recipients. , 1998, Journal of immunology.

[53]  D. Srivastava,et al.  Infusion of cytotoxic T cells for the prevention and treatment of Epstein-Barr virus-induced lymphoma in allogeneic transplant recipients. , 1998, Blood.

[54]  Malcolm K. Brenner,et al.  Long–term restoration of immunity against Epstein–Barr virus infection by adoptive transfer of gene–modified virus–specific T lymphocytes , 1996, Nature Medicine.

[55]  S. Riddell,et al.  Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. , 1995, The New England journal of medicine.

[56]  M. Ladanyi,et al.  Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. , 1994, The New England journal of medicine.

[57]  R. Clark,et al.  Insulin-like growth factor-1 stimulation of lymphopoiesis. , 1993, The Journal of clinical investigation.

[58]  S. Riddell,et al.  Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. , 1992, Science.

[59]  R. Furlanetto,et al.  Insulin-like growth factor-I binds selectively to human peripheral blood monocytes and B-lymphocytes. , 1991, The Journal of clinical endocrinology and metabolism.

[60]  R. Jaenisch,et al.  β2-Microglobulin deficient mice lack CD4−8+ cytolytic T cells , 1990, Nature.

[61]  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.

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