Elsevier

Cellular Immunology

Volume 361, March 2021, 104277
Cellular Immunology

Research paper
Reactive myelopoiesis and the onset of myeloid-mediated immune suppression: Implications for adoptive cell therapy

https://doi.org/10.1016/j.cellimm.2020.104277Get rights and content

Highlights

  • The impact of MDSCs on adoptive T cell therapy is reviewed.

  • Reactive myelopoiesis is driven by lymphodepleting regimens.

  • Strategies to inhibit lymphodepletion-generated MDSCs are discussed.

Abstract

Adoptive T cell therapy (ACT) in combination with lymphodepleting chemotherapy is an effective strategy to induce the eradication of cancer, providing long-term regressions in patients. However, only a minority of patients that receive ACT with tumor-specific T cells exhibit durable benefit. Thus, there is an urgent need to characterize mechanisms of resistance and define strategies to alleviate immunosuppression in the context of ACT in cancer. This article reviews the importance of lymphodepleting regimens in promoting the optimal engraftment and expansion of T cells in hosts after adoptive transfer. In addition, we discuss the role of concomitant immunosuppression and the accumulation of myeloid derived suppressor cells (MDSCs) during immune recovery after lymphodepleting regimens and mobilization regimens.

Introduction

In recent years, there has been an explosion of clinical trials launched to explore the safety and efficacy of ACT for treatment in various malignancies. According to clinicaltrials.gov, more than 300 clinical trials utilizing treatment with lymphodepleting chemotherapies that include cyclophosphamide in combination with ACT are actively recruiting patients. While, preparative lymphodepleting methods are essential to promote the engraftment of adoptively transferred T cells and augment their anti-tumor activity, the concomitant recovery of the endogenous immune system after lymphodepleting regimens can have a profound impact on the function of adoptively transferred T cells. The use of lymphodepleting regimens applies significant pressure on the bone marrow to reconstitute the immune system, which ultimately results in an increased abundance of immunosuppressive myeloid cells. This striking phenomenon is due in part to the mobilization of hematopoietic stem and progenitor cells (HSPCs) from the bone marrow and their differentiation into myeloid cells. The hematopoietic differentiation trajectory and the function of the immune system are hijacked by tumors to promote a growth advantage and evade immune clearance. A variety of factors contribute to the expansion of myeloid cells during stress-induced myelopoiesis which can influence the onset of myeloid-mediated immunosuppression. Tumors preferentially promote the expansion of myeloid cells with potent immunosuppressive functions, including myeloid derived suppressor cells (MDSCs). However, cancer-driven myelopoiesis may differ in the setting of ACT.

In this review, we provide an overview of the known literature pertaining to lymphodepleting regimens that precede ACT. We highlight the resulting accumulation of immunosuppressive myeloid cells which can have profoundly negative consequences on the anti-tumor activity elicited by adoptively transferred T cells. We draw parallels between the known mechanisms that drive myelopoiesis in the presence of pathogenic stimuli or mobilizing cytokines which may also promote the accumulation of myeloid cells in reaction to the stimuli provided by various cytotoxic, lymphodepleting agents. Finally, we provide rationale for strategies to target reactive myelopoiesis with a goal of enhancing therapeutic outcomes for the treatment of cancer with ACT.

Section snippets

Preparative lymphodepletion is essential to elicit durable therapeutic responses to ACT

For nearly 30 years, the infusion of T cells possessing the capability of recognizing and eliminating tumor cells has been explored in patients with cancer. Early studies pioneered by the Rosenberg group at the National Cancer Institute demonstrated that the administration of IL-2 could expand T cells in vivo, leading to durable regressions in patients with metastatic melanoma [1], [2]. This groundbreaking discovery was early evidence that therapies targeting the immune system, but not tumor

Reactive myelopoiesis promotes the expansion of MDSCs

Some of the earliest descriptions of MDSCs in mice and in human subjects were in the context of treatment regimens that included cytokine mobilization or the use of cytotoxic agents, including cyclophosphamide or TBI [29], [30], [31], [32]. The immunosuppressive cells that were described in these studies were broadly characterized as suppressor cells from lymphoid organs after treatment with cyclophosphamide or resembled HSPCs in patients with head and neck cancer [30], [31]. Later studies

The heterogeneity of myeloid cell expansion during reactive myelopoiesis

PMN-MDSCs and M-MDSCs have been shown to be genetically and functionally distinct from physiologic neutrophils and monocytes [72], [73], [74]. Generally, MDSCs have been described to be immunosuppressive, while neutrophils and monocytes from healthy donors or matched hosts do not have an immunosuppressive capacity [72], [74], [75]. However, neutrophils that expand after G-CSF-induced mobilization are phenotypically immature, have a defective chemotactic ability, and exhibit a reduced phagocytic

Factors that drive reactive myelopoiesis in ACT: Opportunity for therapeutic targets?

Indeed, lymphodepleting regimens can deplete immunosuppressive regulatory T cells (Tregs) and MDSCs, but have disparate effects on immune cell reconstitution [101], [102], [103]. Tregs appear to be more sensitive to TBI in comparison to Cy/Flu-based lymphodepletion regimens [103]. Notably, the kinetics of MDSC expansion after lymphodepleting TBI is different than mice treated with chemotherapy. The depletion of MDSCs in TBI-based lymphodepleting regimens is transient and their frequency

The role of danger signals in reactive myelopoiesis

Mounting evidence has highlighted the importance of the microbiota and their products in driving immune responses in cancer [135], [136]. In mice, lymphodepleting doses of cyclophosphamide (i.e. 200 mg/kg body weight in a mouse) can disrupt the gut-epithelial barrier, promoting dysbiosis and the translocation of bacteria from the gut [137]. The efficacy of ACT and effector T cell responses in mice pre-conditioned with cyclophosphamide and antibiotics are reduced in comparison to mice without

Conclusions and future perspectives

Many studies in preclinical models have not included lymphodepleting regimens when studying the anti-tumor effect of adoptively transferred T cells. Indeed, providing an excess of tumor-specific T cells to a tumor-bearing host without any pre-conditioning holds significant value in the investigation of native interactions between cancer and the adaptive immune system. However, the true test of the ability of adoptively transferred T cells to control tumor growth must combine ACT with

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This work was funded by a Bankhead Coley Cancer Research Grant for the Florida Department of Health.

References (164)

  • A.D. Panopoulos et al.

    STAT3 governs distinct pathways in emergency granulopoiesis and mature neutrophils

    Blood

    (2006)
  • I. Marigo et al.

    Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor

    Immunity

    (2010)
  • H.J. Kwak et al.

    Myeloid cell-derived reactive oxygen species externally regulate the proliferation of myeloid progenitors in emergency granulopoiesis

    Immunity

    (2015)
  • G.J. Lieschke et al.

    Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization

    Blood

    (1994)
  • L. Strauss et al.

    RORC1 Regulates Tumor-Promoting “Emergency” Granulo-Monocytopoiesis

    Cancer cell

    (2015)
  • P. Innamarato et al.

    Reactive Myelopoiesis Triggered by Lymphodepleting Chemotherapy Limits the Efficacy of Adoptive T Cell Therapy

    Molecular Therapy

    (2020)
  • A. Luyckx et al.

    G-CSF stem cell mobilization in human donors induces polymorphonuclear and mononuclear myeloid-derived suppressor cells

    Clin Immunol

    (2012)
  • M. Mielcarek et al.

    Suppression of Alloantigen-Induced T-Cell Proliferation by CD14+ Cells Derived From Granulocyte Colony-Stimulating Factor-Mobilized Peripheral Blood Mononuclear Cells

    Blood

    (1997)
  • M. Mielcarek et al.

    Production of Interleukin-10 by Granulocyte Colony-Stimulating Factor-Mobilized Blood Products: A Mechanism for Monocyte-Mediated Suppression of T-Cell Proliferation

    Blood

    (1998)
  • J. Tanaka et al.

    Impaired Induction of the CD28-Responsive Complex in Granulocyte Colony-Stimulating Factor Mobilized CD4 T Cells

    Blood

    (1998)
  • M. Arpinati et al.

    Granulocyte-colony stimulating factor mobilizes T helper 2-inducing dendritic cells

    Blood

    (2000)
  • D.W. Lee et al.

    T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial

    The Lancet

    (2015)
  • A. Vendramin et al.

    Graft Monocytic Myeloid-Derived Suppressor Cell Content Predicts the Risk of Acute Graft-versus-Host Disease after Allogeneic Transplantation of Granulocyte Colony-Stimulating Factor–Mobilized Peripheral Blood Stem Cells

    Biology of Blood and Marrow Transplantation

    (2014)
  • S.A. Rosenberg et al.

    Observations on the Systemic Administration of Autologous Lymphokine-Activated Killer Cells and Recombinant Interleukin-2 to Patients with Metastatic Cancer

    N Engl J Med

    (1985)
  • J.P. Dutcher et al.

    A phase II study of interleukin-2 and lymphokine-activated killer cells in patients with metastatic malignant melanoma.

    JCO

    (1989)
  • M.E. Dudley

    Cancer Regression and Autoimmunity in Patients After Clonal Repopulation with Antitumor Lymphocytes

    Science

    (2002)
  • M.E. Dudley et al.

    Adoptive Cell Therapy for Patients With Metastatic Melanoma: Evaluation of Intensive Myeloablative Chemoradiation Preparative Regimens

    JCO

    (2008)
  • I. Ramachandran et al.

    Systemic and local immunity following adoptive transfer of NY-ESO-1 SPEAR T cells in synovial sarcoma

    j. immunotherapy cancer

    (2019)
  • S.J. Schuster et al.

    Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma

    N Engl J Med

    (2019)
  • C.J. Turtle et al.

    CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients

    The Journal of clinical investigation

    (2016)
  • C.J. Turtle et al.

    Immunotherapy of non-Hodgkin's lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells

    Sci Transl Med

    (2016)
  • S.L. Goff et al.

    Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion Before Adoptive Transfer of Tumor-Infiltrating Lymphocytes for Patients With Metastatic Melanoma

    JCO

    (2016)
  • M.-T. Little et al.

    History of haematopoietic stem-cell transplantation

    Nat Rev Cancer

    (2002)
  • L. Gattinoni et al.

    Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells

    The Journal of experimental medicine

    (2005)
  • C.B. Johnson et al.

    Enhanced Lymphodepletion Is Insufficient to Replace Exogenous IL2 or IL15 Therapy in Augmenting the Efficacy of Adoptively Transferred Effector CD8 + T Cells

    Cancer Res

    (2018)
  • C.B. Johnson et al.

    Effector CD8+ T-cell Engraftment and Antitumor Immunity in Lymphodepleted Hosts Is IL7Ralpha Dependent

    Cancer immunology research

    (2015)
  • D.J. Neitzke et al.

    Murine Th17 cells utilize IL-2 receptor gamma chain cytokines but are resistant to cytokine withdrawal-induced apoptosis

    Cancer Immunol Immunother

    (2017)
  • E.W. Su et al.

    IL-2Ralpha mediates temporal regulation of IL-2 signaling and enhances immunotherapy

    Sci Transl Med

    (2015)
  • M. Cella et al.

    Loss of DNAM-1 contributes to CD8 + T-cell exhaustion in chronic HIV-1 infection : HIGHLIGHTS

    Eur. J. Immunol.

    (2010)
  • J.N. Kochenderfer et al.

    Lymphoma Remissions Caused by Anti-CD19 Chimeric Antigen Receptor T Cells Are Associated With High Serum Interleukin-15 Levels

    JCO

    (2017)
  • C.M. Paulos et al.

    Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling

    J. Clin. Invest.

    (2007)
  • C. Wrzesinski et al.

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

    The Journal of clinical investigation

    (2007)
  • M.J. Besser et al.

    Comprehensive single institute experience with melanoma TIL: Long term clinical results, toxicity profile, and prognostic factors of response

    Mol Carcinog

    (2020)
  • J.E. Mullinax et al.

    Combination of Ipilimumab and Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma

    Frontiers in oncology

    (2018)
  • S. Stevanović et al.

    A Phase II Study of Tumor-infiltrating Lymphocyte Therapy for Human Papillomavirus–associated Epithelial Cancers

    Clin Cancer Res

    (2019)
  • C.M. Suryadevara et al.

    Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma

    OncoImmunology

    (2018)
  • C.M. Bollard et al.

    Tumor-Specific T-Cells Engineered to Overcome Tumor Immune Evasion Induce Clinical Responses in Patients With Relapsed Hodgkin Lymphoma

    JCO

    (2018)
  • M.A. Pule et al.

    Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma

    Nature medicine

    (2008)
  • S. Slavin et al.

    Induction of Allograft Tolerance after Total Lymphoid Irradiation (TLI): Development of Suppressor Cells of the Mixed Leukocyte Reaction (MLR)

    The Journal of Immunology

    (1979)
  • T. Maier et al.

    Murine natural suppressor cells in the newborn, in bone marrow, and after cyclophosphamide

    Genetic variations and dependence on IFN-gamma, The Journal of Immunology

    (1989)
  • View full text