1
|
Dvorakova T, Finisguerra V, Formenti M, Loriot A, Boudhan L, Zhu J, Van den Eynde BJ. Enhanced tumor response to adoptive T cell therapy with PHD2/3-deficient CD8 T cells. Nat Commun 2024; 15:7789. [PMID: 39242595 PMCID: PMC11379939 DOI: 10.1038/s41467-024-51782-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 08/17/2024] [Indexed: 09/09/2024] Open
Abstract
While adoptive cell therapy has shown success in hematological malignancies, its potential against solid tumors is hindered by an immunosuppressive tumor microenvironment (TME). In recent years, members of the hypoxia-inducible factor (HIF) family have gained recognition as important regulators of T-cell metabolism and function. The role of HIF signalling in activated CD8 T cell function in the context of adoptive cell transfer, however, has not been explored in full depth. Here we utilize CRISPR-Cas9 technology to delete prolyl hydroxylase domain-containing enzymes (PHD) 2 and 3, thereby stabilizing HIF-1 signalling, in CD8 T cells that have already undergone differentiation and activation, modelling the T cell phenotype utilized in clinical settings. We observe a significant boost in T-cell activation and effector functions following PHD2/3 deletion, which is dependent on HIF-1α, and is accompanied by an increased glycolytic flux. This improvement in CD8 T cell performance translates into an enhancement in tumor response to adoptive T cell therapy in mice, across various tumor models, even including those reported to be extremely resistant to immunotherapeutic interventions. These findings hold promise for advancing CD8 T-cell based therapies and overcoming the immune suppression barriers within challenging tumor microenvironments.
Collapse
Affiliation(s)
- Tereza Dvorakova
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
- WEL Research Institute, Wavre, 1300, Belgium
| | - Veronica Finisguerra
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
- WEL Research Institute, Wavre, 1300, Belgium
| | - Matteo Formenti
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
- WEL Research Institute, Wavre, 1300, Belgium
| | - Axelle Loriot
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium
| | - Loubna Boudhan
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium
- WEL Research Institute, Wavre, 1300, Belgium
| | - Jingjing Zhu
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium.
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium.
- WEL Research Institute, Wavre, 1300, Belgium.
| | - Benoit J Van den Eynde
- de Duve Institute, UCLouvain, Brussels, B-1200, Belgium.
- Ludwig Institute for Cancer Research, Brussels, B-1200, Belgium.
- WEL Research Institute, Wavre, 1300, Belgium.
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford Oxford, Oxfordshire, UK.
| |
Collapse
|
2
|
Wittling MC, Knochelmann HM, Wyatt MM, Rangel Rivera GO, Cole AC, Lesinski GB, Paulos CM. Distinct host preconditioning regimens differentially impact the antitumor potency of adoptively transferred Th17 cells. J Immunother Cancer 2024; 12:e008715. [PMID: 38945552 PMCID: PMC11216073 DOI: 10.1136/jitc-2023-008715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND How distinct methods of host preconditioning impact the efficacy of adoptively transferred antitumor T helper cells is unknown. METHODS CD4+ T cells with a transgenic T-cell receptor that recognize tyrosinase-related peptide (TRP)-1 melanoma antigen were polarized to the T helper 17 (Th17) phenotype and then transferred into melanoma-bearing mice preconditioned with either total body irradiation or chemotherapy. RESULTS We found that preconditioning mice with a non-myeloablative dose of total body irradiation (TBI of 5 Gy) was more effective than using an equivalently dosed non-myeloablative chemotherapy (cyclophosphamide (CTX) of 200 mg/kg) at augmenting therapeutic activity of antitumor TRP-1 Th17 cells. Antitumor Th17 cells engrafted better following preconditioning with TBI and regressed large established melanoma in all animals. Conversely, only half of mice survived long-term when preconditioned with CTX and infused with anti-melanoma Th17 cells. Interleukin (IL)-17 and interferon-γ, produced by the infused Th17 cells, were detected in animals given either TBI or CTX preconditioning. Interestingly, inflammatory cytokines (granulocyte colony stimulating factor, IL-6, monocyte chemoattractant protein-1, IL-5, and keratinocyte chemoattractant) were significantly elevated in the serum of mice preconditioned with TBI versus CTX after Th17 therapy. The addition of fludarabine (FLU, 200 mg/kg) to CTX (200 mg/kg) improved the antitumor response to the same degree mediated by TBI, whereas FLU alone with Th17 therapy was ineffective. CONCLUSIONS Our results indicate, for the first time, that the antitumor response, persistence, and cytokine profiles resulting from Th17 therapy are impacted by the specific regimen of host preconditioning. This work is important for understanding mechanisms that promote long-lived responses by adoptive cellular therapy, particularly as CD4+ based T-cell therapies are now emerging in the clinic.
Collapse
Affiliation(s)
- Megen C Wittling
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Hannah M Knochelmann
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Megan M Wyatt
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| | - Guillermo O Rangel Rivera
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
- Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anna C Cole
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| | | | - Chrystal M Paulos
- Surgery/Oncology & Microbiology/Immunology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Wittling MC, Knochelmann HM, Wyatt MM, Rangel Rivera GO, Cole AC, Lesinski GB, Paulos CM. Distinct host preconditioning regimens differentially impact the antitumor potency of adoptively transferred Th17 cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572179. [PMID: 38187594 PMCID: PMC10769197 DOI: 10.1101/2023.12.18.572179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Background Mechanisms by which distinct methods of host preconditioning impact the efficacy of adoptively transferred antitumor T helper cells is unknown. Methods CD4 + T cells with a transgenic TCR that recognize TRP-1 melanoma antigen were polarized to the T helper 17 (Th17) phenotype and then transferred into melanoma-bearing mice preconditioned with either total body irradiation or chemotherapy. Results We found that preconditioning mice with a non-myeloablative dose of total body irradiation (TBI of 5 Gy) was more effective than using an equivalently dosed non-myeloablative chemotherapy (CTX at 200 mg/kg) at augmenting therapeutic activity of anti-tumor TRP-1 Th17 cells. Anti-tumor Th17 cells engrafted better following preconditioning with TBI and regressed large established melanoma in all animals. Conversely, only half of mice survived long-term when preconditioned with CTX and infused with anti-melanoma Th17 cells. IL-17 and IFN-g produced by the infused Th17 cells, were detected in animals given either TBI or CTX preconditioning. Interestingly, inflammatory cytokines (G-CSF, IL-6, MCP-1, IL-5, and KC) were significantly elevated in the serum of mice preconditioned with TBI versus CTX after Th17 therapy. Conclusions Our results indicate, for the first time, that the antitumor response, persistence, and cytokine profiles resulting from Th17 therapy are impacted by the specific regimen of host preconditioning. This work is important for understanding mechanisms that promote long-lived responses by ACT, particularly as CD4 + based T cell therapies are now emerging in the clinic.
Collapse
|
4
|
Liu S, Li J, Gu L, Wu K, Xing H. Nanoparticles for Chemoimmunotherapy Against Triple-Negative Breast Cancer. Int J Nanomedicine 2022; 17:5209-5227. [PMID: 36388877 PMCID: PMC9651025 DOI: 10.2147/ijn.s388075] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) exhibits high recurrence and mortality rates because of the lack of effective treatment targets. Surgery and traditional chemotherapy are the primary treatment options. Immunotherapy shows high potential for treating various cancers but exhibits limited efficacy against TNBC as a monotherapy. Chemoimmunotherapy has broad prospects for applications for cancer treatment conferred through the synergistic immunomodulatory and anti-tumor effects of chemotherapy and immunotherapeutic strategies. However, improving the efficacy of synergistic therapy and reducing the side effects of multiple drugs remain to be the main challenges in chemoimmunotherapy against TNBC. Nanocarriers can target both cancer and immune cells, promote drug accumulation, and show minimal toxicity, making them ideal delivery systems for chemotherapeutic and immunotherapeutic agents. In this review, we introduce the immunomodulatory effects of chemotherapy and combined mechanisms of chemoimmunotherapy, followed by a summary of nanoparticle-mediated chemoimmunotherapeutic strategies used for treating TNBC. This up-to-date synthesis of relevant findings in the field merits contemplation, while considering avenues of investigation to enable advances in the field.
Collapse
Affiliation(s)
- Siyan Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Jing Li
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Lin Gu
- Breast Surgery, Jilin Province Tumor Hospital, Changchun, People’s Republic of China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Hua Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
5
|
Anti-Gr-1 Antibody Provides Short-Term Depletion of MDSC in Lymphodepleted Mice with Active-Specific Melanoma Therapy. Vaccines (Basel) 2022; 10:vaccines10040560. [PMID: 35455309 PMCID: PMC9032646 DOI: 10.3390/vaccines10040560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/28/2022] Open
Abstract
Lymphodepletion, reconstitution and active-specific tumor cell vaccination (LRAST) enhances the induction of tumor-specific T cells in a murine melanoma model. Myeloid-derived suppressor cells (MDSC) may counteract the induction of tumor-reactive T cells and their therapeutic efficacy. Thus, the aim of the study was to evaluate a possible benefit of MDSC depletion using anti-Gr-1 antibodies (Ab) in combination with LRAST. Female C57BL/6 mice with 3 days established subcutaneous (s.c.) D5 melanoma were lymphodepleted with cyclophosphamide and reconstituted with naive splenocytes. Vaccination was performed with irradiated syngeneic mGM-CSF-secreting D5G6 melanoma cells. MDSC depletion was performed using anti-Gr-1 Ab (clone RB6-8C5). Induction of tumor-specific T cells derived from tumor vaccine draining lymph nodes (TVDLN) was evaluated by the amount of tumor-specific interferon (IFN)-γ release. LRAST combined with anti-Gr-1 mAb administration enhanced the induction of tumor-specific T cells in TVDLN capable of releasing IFN-γ in a tumor-specific manner. Additional anti-Gr-1 mAb administration in LRAST-treated mice delayed growth of D5 melanomas by two weeks. Furthermore, we elucidate the impact of anti-Gr-1-depleting antibodies on the memory T cell compartment. Our data indicate that standard of care treatment regimens against cancer can be improved by implementing agents, e.g., depleting antibodies, which target and eliminate MDSC.
Collapse
|
6
|
Proskurina AS, Ruzanova VS, Ostanin AA, Chernykh ER, Bogachev SS. Theoretical premises of a "three in one" therapeutic approach to treat immunogenic and nonimmunogenic cancers: a narrative review. Transl Cancer Res 2022; 10:4958-4972. [PMID: 35116346 PMCID: PMC8797664 DOI: 10.21037/tcr-21-919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Objective We describe experimental and theoretical premises of a powerful cancer therapy based on the combination of three approaches. These include (I) in situ vaccination (intratumoral injections of CpG oligonucleotides and anti-OX40 antibody); (II) chronometric or metronomic low-dose cyclophosphamide (CMLD CP)-based chemotherapy; (III) cancer stem cell-eradicating therapy referred to as Karanahan (from the Sanskrit kāraṇa [“source”] + han [“to kill”]). Background In murine models, the first two approaches are particularly potent in targeting immunogenic tumors for destruction. In situ vaccination activates a fully fledged anticancer immune response via an intricate network of ligand–receptor–cytokine interactions. CMLD CP-based chemotherapy primarily targets the suppressive tumor microenvironment and activates tumor-infiltrating effectors. In contrast, Karanahan technology, being aimed at replicative machinery of tumor cells (both stem-like and committed), does not depend on tumor immunogenicity. With this technology, mice engrafted with ascites and/or solid tumors can be successfully cured. There is a significant degree of mechanistic and therapeutic overlap between these three approaches. For instance, the similarities shared between in situ vaccination and Karanahan technology include the therapeutic procedure, the cell target [antigen-presenting cells (APC) and dendritic cells (DC)], and the use of DNA-based preparations (CpG and DNAmix). Features shared between CMLD CP-based chemotherapy and Karanahan technology are the timing and the dose of the cytostatic drug administration, which lead to tumor regression. Methods The following keywords were used to search PubMed for the latest research reporting successful eradication of transplantable cancers in animal models that relied on approaches distinct from those used in the Karanahan technology: eradication of malignancy, cure cancer, complete tumor regression, permanently eradicating advanced mouse tumor, metronomic chemotherapy, in situ vaccination, immunotherapy, and others. Conclusion We hypothesize, therefore, that very potent anticancer activity can be achieved once these three therapeutic modalities are combined into a single approach. This multimodal approach is theoretically curative for any type of cancer that depends on the presence of tumor-inducing cancer stem cells, provided that the active therapeutic components are efficiently delivered into the tumor and the specific biological features of a given patient’s tumor are properly addressed. We expect this multimodal approach to be primarily applicable to late-stage or terminal cancer patients who have exhausted all treatment options as well as patients with inoperable tumors.
Collapse
Affiliation(s)
- Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Vera S Ruzanova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | - Alexandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
7
|
Ciccolella M, Andreone S, Mancini J, Sestili P, Negri D, Pacca AM, D’Urso MT, Macchia D, Canese R, Pang K, SaiYing Ko T, Decadt Y, Schiavoni G, Mattei F, Belardelli F, Aricò E, Bracci L. Anticancer Effects of Sublingual Type I IFN in Combination with Chemotherapy in Implantable and Spontaneous Tumor Models. Cells 2021; 10:845. [PMID: 33917958 PMCID: PMC8068355 DOI: 10.3390/cells10040845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023] Open
Abstract
Salivary gland tumors are a heterogeneous group of neoplasms representing less than 10% of all head and neck tumors. Among salivary gland tumors, salivary duct carcinoma (SDC) is a rare, but highly aggressive malignant tumor resembling ductal breast carcinoma. Sublingual treatments are promising for SDC due to the induction of both local and systemic biological effects and to reduced systemic toxicity compared to other administration routes. In the present study, we first established that the sublingual administration of type I IFN (IFN-I) is safe and feasible, and exerts antitumor effects both as monotherapy and in combination with chemotherapy in transplantable tumor models, i.e., B16-OVA melanoma and EG.7-OVA lymphoma. Subsequently, we proved that sublingual IFN-I in combination with cyclophosphamide (CTX) induces a long-lasting reduction of tumor mass in NeuT transgenic mice that spontaneously develop SDC. Most importantly, tumor shrinkage in NeuT transgenic micewas accompanied by the emergence of tumor-specific cellular immune responses both in the blood and in the tumor tissue. Altogether, these results provide evidence that sublingual IFN holds promise in combination with chemotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Maria Ciccolella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Jacopo Mancini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Paola Sestili
- National Center for the Control and Evaluation of Medicines, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anna Maria Pacca
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Maria Teresa D’Urso
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Daniele Macchia
- Animal Research and Welfare Centre, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.M.P.); (M.T.D.); (D.M.)
| | - Rossella Canese
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Ken Pang
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
- Murdoch Children’s Research Institute, Parkville 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville 3010, Australia
| | - Thomas SaiYing Ko
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Yves Decadt
- Biolingus AG, CH-6052 Hergiswil NW, Switzerland; (K.P.); (T.S.K.); (Y.D.)
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| | - Filippo Belardelli
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 00133 Rome, Italy;
| | - Eleonora Aricò
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.C.); (S.A.); (J.M.); (G.S.); (F.M.)
| |
Collapse
|
8
|
Jia Q, Qin D, He F, Xie Q, Ying Z, Zhang Y, Song Y, Cheng JN, Zuo X, Xu L, Fang H, Hu C, Peng L, Jin T, Shi Z, Alexander PB, Wang Y, Liu Y, Han W, Zhu J, Wang P, Li QJ, Zhu B. Peripheral eosinophil counts predict efficacy of anti-CD19 CAR-T cell therapy against B-lineage non-Hodgkin lymphoma. Am J Cancer Res 2021; 11:4699-4709. [PMID: 33754022 PMCID: PMC7978305 DOI: 10.7150/thno.54546] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/14/2021] [Indexed: 02/05/2023] Open
Abstract
Rationale: The onset of cytokine release syndrome (CRS) and in vivo persistence of anti-CD19 chimeric antigen receptor T (CAR-T) cells after infusion correlate with clinical responsiveness. However, there are no known baseline biomarkers that can predict the prognosis of patients with B-lineage non-Hodgkin lymphoma (B-NHL). The aim of this study was to identify blood cell populations associated with beneficial outcomes in B-NHL patients administered CAR-T cell immunotherapies. Methods: We enumerated peripheral blood and CAR-T cells by retrospectively analyzing three CAR-T cell trials involving 65 B-NHL patients. We used a preclinical model to elucidate the eosinophil mechanism in CAR-T cell therapy. Results: During an observation period up to 30 mo, B-NHL patients with higher baseline eosinophil counts had higher objective response rates than those with low eosinophil counts. Higher baseline eosinophil counts were also significantly associated with durable progression-free survival (PFS). The predictive significance of baseline eosinophil counts was validated in two independent cohorts. A preclinical model showed that eosinophil depletion impairs the intratumoral infiltration of transferred CAR-T cells and reduces CAR-T cell antitumor efficacy. Conclusion: The results of this study suggest that peripheral eosinophils could serve as stratification biomarkers and a recruitment machinery to facilitate anti-CD19 CAR-T cell therapy in B-NHL patients.
Collapse
|
9
|
Slingluff CL, Petroni GR, Chianese-Bullock KA, Wages NA, Olson WC, Smith KT, Haden K, Dengel LT, Dickinson A, Reed C, Gaughan EM, Grosh WW, Kaur V, Varhegyi N, Smolkin M, Galeassi NV, Deacon D, Hall EH. Trial to evaluate the immunogenicity and safety of a melanoma helper peptide vaccine plus incomplete Freund's adjuvant, cyclophosphamide, and polyICLC (Mel63). J Immunother Cancer 2021; 9:jitc-2020-000934. [PMID: 33479025 PMCID: PMC7825263 DOI: 10.1136/jitc-2020-000934] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2020] [Indexed: 12/17/2022] Open
Abstract
Background Peptide vaccines designed to stimulate melanoma-reactive CD4+ T cells can induce T cell and antibody (Ab) responses, associated with enhanced overall survival. We hypothesized that adding toll-like receptor 3 agonist polyICLC to an incomplete Freund’s adjuvant (IFA) would be safe and would support strong, durable CD4+ T cell and Ab responses. We also hypothesized that oral low-dose metronomic cyclophosphamide (mCy) would be safe, would reduce circulating regulatory T cells (T-regs) and would further enhance immunogenicity. Participants and methods An adaptive design based on toxicity and durable CD4+ T cell immune response (dRsp) was used to assign participants with resected stage IIA-IV melanoma to one of four study regimens. The regimens included a vaccine comprising six melanoma peptides restricted by Class II MHC (6MHP) in an emulsion with IFA alone (Arm A), with IFA plus systemic mCy (Arm B), with IFA+ local polyICLC (Arm C), or with IFA+ polyICLC+ mCy (Arm D). Toxicities were recorded (CTCAE V.4.03). T cell responses were measured by interferon γ ELIspot assay ex vivo. Serum Ab responses to 6MHP were measured by ELISA. Circulating T-regs were assessed by flow cytometry. Results Forty-eight eligible participants were enrolled and treated. Early data on safety and dRsp favored enrollment on arm D. Total enrollment on Arms A-D were 3, 7, 6, and 32, respectively. Treatment-related dose-limiting toxicities (DLTs) were observed in 1/7 (14%) participants on arm B and 2/32 (6%) on arm D. None exceeded the 25% DLT threshold for early closure to enrollment for any arm. Strong durable T cell responses to 6MHP were detected ex vivo in 0%, 29%, 67%, and 47% of participants on arms A-D, respectively. IgG Ab responses were greatest for arms C and D. Circulating T-regs frequencies were not altered by mCy. Conclusions 6MHP vaccines administered with IFA, polyICLC, and mCy were well tolerated. The dRsp rate for arm D of 47% (90% CI 32 to 63) exceeded the 18% (90% CI 11 to 26) rate previously observed with 6MHP in IFA alone. Vaccination with IFA+ polyICLC (arm C) also showed promise for enhancing T cell and Ab responses.
Collapse
Affiliation(s)
- Craig L Slingluff
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA .,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Gina R Petroni
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Nolan A Wages
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Walter C Olson
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kelly T Smith
- Office of Research Cores Administration, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kathleen Haden
- University of Virginia Cancer Center, Charlottesville, Virginia, USA.,University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Lynn T Dengel
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Anna Dickinson
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Caroline Reed
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Elizabeth M Gaughan
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - William W Grosh
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Varinder Kaur
- Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nikole Varhegyi
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark Smolkin
- Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nadejda V Galeassi
- Cardiovascular Imaging Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Donna Deacon
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emily H Hall
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,University of Virginia Cancer Center, Charlottesville, Virginia, USA
| |
Collapse
|
10
|
Innamarato P, Pilon-Thomas S. Reactive myelopoiesis and the onset of myeloid-mediated immune suppression: Implications for adoptive cell therapy. Cell Immunol 2020; 361:104277. [PMID: 33476931 DOI: 10.1016/j.cellimm.2020.104277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/17/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Patrick Innamarato
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shari Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
11
|
Ring SS, Królik M, Hartmann F, Schmidt E, Ali OH, Ludewig B, Kochanek S, Flatz L. Heterologous Prime Boost Vaccination Induces Protective Melanoma-Specific CD8 + T Cell Responses. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:179-187. [PMID: 33209978 PMCID: PMC7658660 DOI: 10.1016/j.omto.2020.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022]
Abstract
Cancer vaccination aims at inducing an adaptive immune response against tumor-derived antigens. In this study, we utilize recombinant human adenovirus serotype 5 (rAd5) and recombinant lymphocytic choriomeningitis virus (rLCMV)-based vectors expressing the melanocyte differentiation antigen gp100. In contrast to single or homologous vaccination, a heterologous prime boost vaccination starting with a rAd5-gp100 prime immunization followed by a rLCMV-gp100 boost injection induces a high magnitude of polyfunctional gp100-specific CD8+ T cells. Our data indicate that an optimal T cell induction is dependent on the order and interval of the vaccinations. A prophylactic prime boost vaccination with rAd5- and rLCMV-gp100 protects mice from a B16.F10 melanoma challenge. In the therapeutic setting, combination of the vaccination with low-dose cyclophosphamide showed a synergistic effect and significantly delayed tumor growth. Our findings suggest that heterologous viral vector prime boost immunizations can mediate tumor control in a mouse melanoma model.
Collapse
Affiliation(s)
- Sandra S Ring
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Michał Królik
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Fabienne Hartmann
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| | - Erika Schmidt
- Department of Gene Therapy, Ulm University, Helmholtzstrasse 8, 89081 Ulm, Germany
| | - Omar Hasan Ali
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Stefan Kochanek
- Department of Gene Therapy, Ulm University, Helmholtzstrasse 8, 89081 Ulm, Germany
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Department of Dermatology, University Hospital Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.,Department of Oncology and Hematology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland.,Department of Dermatology, Kantonsspital St.Gallen, Rorschacher Strasse 95, 9007 St.Gallen, Switzerland
| |
Collapse
|
12
|
Delahousse J, Skarbek C, Desbois M, Perfettini JL, Chaput N, Paci A. Oxazaphosphorines combined with immune checkpoint blockers: dose-dependent tuning between immune and cytotoxic effects. J Immunother Cancer 2020; 8:jitc-2020-000916. [PMID: 32784216 PMCID: PMC7418776 DOI: 10.1136/jitc-2020-000916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 11/18/2022] Open
Abstract
Background Oxazaphosphorines (cyclophosphamide (CPA), ifosfamide (IFO)) are major alkylating agents of polychemotherapy protocols but limiting their toxicity and increasing their efficacy could be of major interest. Oxazaphosphorines are prodrugs that require an activation by cytochrome P450 (CYP). CPA is mainly metabolized (>80%) to phosphoramide mustard while only 10%–50% of IFO is transformed in the alkylating entity, isophosphoramide mustard and 50%–90% of IFO release chloroacetaldehyde, a nephrotoxic and neurotoxic metabolite. Geranyloxy-IFO (G-IFO) was reported as a preactivated IFO to circumvent the toxic pathway giving directly the isophosphoramide mustard without CYP metabolization. The similarity in structure of CPA and IFO and the similarity in metabolic balance of CPA and G-IFO have led us to explore immunomodulatory effect of these components in mice and to investigate the combination of these oxazaphosphorines with immune checkpoint blockers (ICB). Methods The investigation of the immunomodulatory properties of IFO and G-IFO compared with CPA has been conducted through immune cell phenotyping by flow cytometry and analysis of the cytokine profile of T cells after ex-vivo restimulation. T cell-mediated antitumor efficacy was confirmed in CD4+ and CD8+ T cell-depleted mice. A combination of oxazaphosphorines with an anti-programmed cell death 1 (PD-1) antibody has been studied in MCA205 tumor-bearing mice. Results Studies on a MCA205 mouse model have demonstrated a dose-dependent effect of IFO and G-IFO on T cell immunity. These components in particular favored Th1 polarization when used at low dose (150 and eq. 100 mg/kg, respectively). Antitumor activity at low dose was abolished in mice depleted in CD4+ and CD8+ T cells. G-IFO at low dose (eq. 100 mg/kg) in combination with anti-PD-1 antidody showed high synergistic antitumor efficacy compared with IFO. Conclusion Oxazaphosphorines are characterized by a dual mechanism of antitumor action; low-dose schedules should be preferred in combination with ICB, and dose escalation was found to have better utility in polychemotherapy protocols where a conventional direct cytotoxic anticancer effect is needed. G-IFO, the novel oxazaphosphorine drug, has shown a better metabolic index compared with IFO as its metabolization gives mainly the alkylating mustard as CPA (and not IFO) and a best potential in combination with ICB.
Collapse
Affiliation(s)
- Julia Delahousse
- Molecular Radiotherapy and Innovative Therapeutics, Unité Mixte de Recherche 1030 INSERM, Gustave Roussy, F-94805, Villejuif, France.,Vectorology and Anticancer Therapies, Unité Mixte de Recherche 8203 Centre National de la Recherche Scientifique, Gustave Roussy, F-94805, Villejuif, France
| | - Charles Skarbek
- Vectorology and Anticancer Therapies, Unité Mixte de Recherche 8203 Centre National de la Recherche Scientifique, Gustave Roussy, F-94805, Villejuif, France
| | - Mélanie Desbois
- Laboratoire d'immunomonitoring En Oncologie, Gustave Roussy, F-94805, Villejuif, France
| | - Jean-Luc Perfettini
- Molecular Radiotherapy and Innovative Therapeutics, Unité Mixte de Recherche 1030 INSERM, Gustave Roussy, F-94805, Villejuif, France
| | - Nathalie Chaput
- Laboratoire d'immunomonitoring En Oncologie, Gustave Roussy, F-94805, Villejuif, France.,Laboratory of Genetic Instability and Oncogenesis, Unité Mixte de Recherche 8200 Centre National de la Recherche Scientifique, Gustave Roussy Institute, F-94805, Villejuif, France.,Faculté de Pharmacie, Université Paris-Saclay, F-92296, Chatenay-Malabry, France
| | - Angelo Paci
- Molecular Radiotherapy and Innovative Therapeutics, Unité Mixte de Recherche 1030 INSERM, Gustave Roussy, F-94805, Villejuif, France .,Faculté de Pharmacie, Université Paris-Saclay, F-92296, Chatenay-Malabry, France.,Pharmacology Department, Gustave Roussy, Villejuif, France
| |
Collapse
|
13
|
Walsh SR, Simovic B, Chen L, Bastin D, Nguyen A, Stephenson K, Mandur TS, Bramson JL, Lichty BD, Wan Y. Endogenous T cells prevent tumor immune escape following adoptive T cell therapy. J Clin Invest 2020; 129:5400-5410. [PMID: 31682239 DOI: 10.1172/jci126199] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
While the outcome of adoptive T cell therapy (ACT) is typically correlated with the functionality of the inoculated T cells, the role of the endogenous T cells is unknown. The success of checkpoint blockade therapy has demonstrated the potentially curative value of preexisting tumor-primed T cells in cancer treatment. Given the results from checkpoint blockade therapy, we hypothesized that endogenous T cells contribute to long-term survival following ACT. Here, we describe a therapeutic approach combining ACT with an oncolytic vaccine that allows simultaneous analysis of antitumor immunity mediated by transferred and endogenous T cells. We found that, in addition to promoting the expansion and tumor infiltration of the transferred T cells, oncolytic vaccines boosted tumor-primed host T cells. We determined that transferred T cells contributed to rapid destruction of large tumor masses while endogenous T cells concurrently prevented the emergence of antigen-loss variants. Moreover, while transferred T cells disappeared shortly after tumor regression, endogenous T cells secured long-term memory with a broad repertoire of antigen specificity. Our findings suggest that this combination strategy may exploit the full potential of ACT and tumor-primed host T cells to eliminate the primary tumor, prevent immune escape, and provide long-term protective memory.
Collapse
|
14
|
Escribà-Garcia L, Alvarez-Fernández C, Caballero AC, Schaub R, Sierra J, Briones J. The novel agonistic iNKT-cell antibody NKT14m induces a therapeutic antitumor response against B-cell lymphoma. Oncoimmunology 2019; 8:e1546543. [PMID: 30713807 DOI: 10.1080/2162402x.2018.1546543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 02/02/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a small population of T lymphocytes that expresses an invariant T cell receptor with a unique specificity for glycolipid antigens. Their activation using the glycolipid α-galactosylceramide (α-GalCer) triggers innate and adaptive immune responses. The use of α-GalCer in preclinical models as a single antitumor treatment showed moderate effect, but its efficacy in cancer patients was less effective. In addition, this glycolipid induces long-term iNKT-cell anergy precluding the possibility of retreatment. Recently, the first murine iNKT-cell agonistic antibody, NKT14m, has been developed. Here, we analyzed, for the first time, the antitumor efficacy of NKT14m in a B-cell lymphoma model. In a therapeutic setting, a single dose of NKT14m had a moderate antitumor efficacy that was associated with an increase of IFN-γ producing iNKT cells even after a second dose of the NKT14m antibody. Importantly, the combination of a single dose of NKT14m with cyclophosphamide had a potent antitumor efficacy and long-lasting immunity in vivo. Our findings provide the first evidence of the in vivo antitumor efficacy of NKT14m antibody, showing that, either alone or in combination with chemotherapy, induces an effective antitumor response. These results open new opportunities for iNKT-cell mediated immunotherapy to treat B-cell lymphoma.
Collapse
Affiliation(s)
- Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| |
Collapse
|
15
|
Rodríguez Bustos H, Espinoza-Navarro O, Castro ME. Effect of classical conditioning over immature and mature B lymphocytes regulation. Microsc Res Tech 2018; 81:1451-1455. [PMID: 30365190 DOI: 10.1002/jemt.23112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 06/11/2018] [Accepted: 07/23/2018] [Indexed: 11/09/2022]
Abstract
Classical conditioning (CC) with a sweet flavored is useful for experimental models of immune response. The objective this work was analyze spleen cell response on the differentiation of B-lymphocytes associated with the CC with sweet flavored in adult rats. Twelve adult male rats were divided (n = 3) in Group 1 (control); Group 2 without conditioned stimulus (WCS) received only water; Group 3 receiving only water with salt; Group 4 (experimental) with CS of sweet flavored and salty flavored afterwards. In every work group, all animals were subjected to the unconditioned stimulus (US) treated by 10 mg/kg body weight of cyclophosphamide injected intraperitoneally (i.p.). Subsequently an immunological challenge with sheep red blood cells i.p. was applied. Spleen samples were obtained 15 days after using euthanasia with 1% sodium thiopental i.p. The evaluation B lymphocyte (immature and mature) was performed by immunohistochemistry using specific antibodies against CD20 and against CD37, and revealed with HRP/DAB. The results show in every work group significant increases of CD20+ and CD37+ cell densities in the red pulp of the spleen of male adult rats from different groups of the experimental design. We concluded that the immunomodulatory response under CC might facilitate a less aggressive and more physiological immunological response against immunosuppression. RESEARCH HIGHLIGHTS: The immune response can be reinforced by classical conditioning. The sweet taste allows to positively condition the immune response. B lymphocytes actively participate in the immune response and classical conditioning.
Collapse
Affiliation(s)
- Héctor Rodríguez Bustos
- Laboratory of Morphology, Anatomy and Development of Biology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Omar Espinoza-Navarro
- Laboratory of Morphology, Anatomy and Development of Biology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María Eugenia Castro
- Laboratory of Morphology, Anatomy and Development of Biology, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
16
|
Hughes E, Scurr M, Campbell E, Jones E, Godkin A, Gallimore A. T-cell modulation by cyclophosphamide for tumour therapy. Immunology 2018; 154:62-68. [PMID: 29460448 PMCID: PMC5904691 DOI: 10.1111/imm.12913] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/14/2022] Open
Abstract
The power of T cells for cancer treatment has been demonstrated by the success of co-inhibitory receptor blockade and adoptive T-cell immunotherapies. These treatments are highly successful for certain cancers, but are often personalized, expensive and associated with harmful side effects. Other T-cell-modulating drugs may provide additional means of improving immune responses to tumours without these disadvantages. Conventional chemotherapeutic drugs are traditionally used to target cancers directly; however, it is clear that some also have significant immune-modulating effects that can be harnessed to target tumours. Cyclophosphamide is one such drug; used at lower doses than in mainstream chemotherapy, it can perturb immune homeostasis, tipping the balance towards generation of anti-tumour T-cell responses and control of cancer growth. This review discusses its growing reputation as an immune-modulator whose multiple effects synergize with the microbiota to tip the balance towards tumour immunity offering widespread benefits as a safe, and relatively inexpensive component of cancer immunotherapy.
Collapse
Affiliation(s)
- Ellyn Hughes
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
- Present address:
Faculty of Medicine Nursing and Health SciencesSchool of Biomedical SciencesMonash UniversityMelbourneAustralia
| | - Martin Scurr
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
| | - Emma Campbell
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
| | - Emma Jones
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
| | - Andrew Godkin
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
| | - Awen Gallimore
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityCardiffUK
| |
Collapse
|
17
|
Salem ML, El-Naggar SA, Mahmoud HA, Elgharabawy RM, Bader AM. Cyclophosphamide eradicates murine immunogenic tumor coding for a non-self-antigen and induces antitumor immunity. Int J Immunopathol Pharmacol 2018; 32:2058738418796591. [PMID: 30270681 PMCID: PMC6168726 DOI: 10.1177/2058738418796591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Although the majority of cancers respond to chemotherapy, most cancer types relapse, at least in part, due to the poor immunogenicity of most tumor. We have reported before that treatment of tumor bearing mice with a combination of the anti-cancer chemotherapy cyclophosphamide (CTX) and immunotherapy can result in complete tumor regression using T-cell receptor (TCR) transgenic CD8+ T cells specific to antigens. This study aimed to determine whether chemotherapy can cure immunogenic tumor which expresses non-self-tumor antigen and result in antitumor immunity. Either EL4 cell line, a poorly immunogenic thymoma, or EG7, a clone of EL4 cells transfected with ovalbumin (OVA), as a non-self-antigen were inoculated subcutaneously into wild type or splenectomized C57BL/6 mice and then treated once with intraperitoneal (i.p.) injection of 4 mg CTX/mouse. In certain experiments, the mice were rechallenged with the same tumor type 1-2 months after the primary challenge. Treatment of EL4 bearing mice with CTX induced transient antitumor effect followed by tumor progression. Interestingly, however, treatment of EG7-bearing mice with CTX resulted in regression of early and advanced tumors. EG7 tumor-free mice rejected the second and the third challenges with EG7 cells, but not with challenge EL4 cells. These antitumor effects did not require spleen, since splenectomized mice showed similar antitumor effects of CTX on EG7 cells. Taken together, these data indicate that expression of non-self-antigen by poorly immunogenic tumor might be a reliable means to increase its immunogenicity and its response to chemotherapy.
Collapse
Affiliation(s)
- Mohamed L Salem
- Immunology and Biotechnology Unit,
Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
- Center of Excellence in Cancer Research,
New Teaching Hospital, Tanta University, Tanta, Egypt
- Mohamed L Salem, Immunology and
Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University,
Tanta, Egypt. ;
| | - Sabry A El-Naggar
- Immunology and Biotechnology Unit,
Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
- Center of Excellence in Cancer Research,
New Teaching Hospital, Tanta University, Tanta, Egypt
| | - Heba A Mahmoud
- Department of Pharmacology, College of
Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rehab M Elgharabawy
- Pharmacology and Toxicology Department,
Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Abeer M Bader
- Zoology Department, Faculty of Science,
Cairo University, Giza, Egypt
| |
Collapse
|
18
|
Gebremeskel S, Lobert L, Tanner K, Walker B, Oliphant T, Clarke LE, Dellaire G, Johnston B. Natural Killer T-cell Immunotherapy in Combination with Chemotherapy-Induced Immunogenic Cell Death Targets Metastatic Breast Cancer. Cancer Immunol Res 2017; 5:1086-1097. [PMID: 29054890 DOI: 10.1158/2326-6066.cir-17-0229] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/11/2017] [Accepted: 10/13/2017] [Indexed: 11/16/2022]
Abstract
Natural killer T (NKT) cells are glycolipid-reactive lymphocytes that promote cancer control. In previous studies, NKT-cell activation improved survival and antitumor immunity in a postsurgical mouse model of metastatic breast cancer. Herein, we investigated whether NKT-cell activation could be combined with chemotherapeutic agents to augment therapeutic outcomes. Gemcitabine and cyclophosphamide analogues enhanced the potential immunogenicity of 4T1 mammary carcinoma cells by increasing the expression of antigen-presenting molecules (MHC-I, MHC-II, and CD1d) and promoting exposure or release of immunogenic cell death markers (calreticulin, HMGB1, and ATP). In 4T1 primary tumor and postsurgical metastasis models, BALB/c mice were treated with cyclophosphamide or gemcitabine. NKT cells were then activated by transfer of dendritic cells loaded with the glycolipid antigen α-galactosylceramide (α-GalCer). Chemotherapeutic treatments did not impact NKT-cell activation but enhanced recruitment into primary tumors. Cyclophosphamide, gemcitabine, or α-GalCer-loaded dendritic cell monotherapies decreased tumor growth in the primary tumor model and reduced metastatic burden and prolonged survival in the metastasis model. Combining chemotherapeutics with NKT-cell activation therapy significantly enhanced survival, with surviving mice exhibiting attenuated tumor growth following a second tumor challenge. The frequency of myeloid-derived suppressor cells was reduced by gemcitabine, cyclophosphamide, or α-GalCer-loaded dendritic cell treatments; cyclophosphamide also reduced the frequency of regulatory T cells. Individual treatments increased immune cell activation, cytokine polarization, and cytotoxic responses, although these readouts were not enhanced further by combining therapies. These findings demonstrate that NKT-cell activation therapy can be combined with gemcitabine or cyclophosphamide to target tumor burden and enhance protection against tumor recurrence. Cancer Immunol Res; 5(12); 1086-97. ©2017 AACR.
Collapse
Affiliation(s)
- Simon Gebremeskel
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Lynnea Lobert
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada.,Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Kaitlyn Tanner
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brynn Walker
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tora Oliphant
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Livia E Clarke
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Graham Dellaire
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Brent Johnston
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada. .,Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.,Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
19
|
Nejati R, Goldstein JB, Halperin DM, Wang H, Hejazi N, Rashid A, Katz MH, Lee JE, Fleming JB, Rodriguez-Canales J, Blando J, Wistuba II, Maitra A, Wolff RA, Varadhachary GR, Wang H. Prognostic Significance of Tumor-Infiltrating Lymphocytes in Patients With Pancreatic Ductal Adenocarcinoma Treated With Neoadjuvant Chemotherapy. Pancreas 2017; 46:1180-1187. [PMID: 28902789 PMCID: PMC5790553 DOI: 10.1097/mpa.0000000000000914] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The aim of this study was to examine tumor-infiltrating lymphocytes (TILs) and their prognostic value in patients with pancreatic ductal adenocarcinoma (PDAC) after neoadjuvant therapy. METHODS Intratumoral CD4, CD8, and FOXP3 lymphocytes were examined by immunohistochemistry using a computer-assisted quantitative analysis in 136 PDAC patients who received neoadjuvant therapy and pancreaticoduodenectomy. The results were correlated with clinicopathological parameters and survival. RESULTS High CD4 TILs in treated PDAC were associated with high CD8 TILs (P = 0.003), differentiation (P = 0.04), and a lower frequency of recurrence (P = 0.02). Patients with high CD4 TILs had longer disease-free survival and overall survival (OS) than did patients with low CD4 TILs (P < 0.01). The median OS of patients with a high CD8/FOXP3 lymphocyte ratio (39.5 [standard deviation, 6.1] months) was longer than that of patients with a low CD8/FOXP3 lymphocyte ratio (28.3 [standard deviation, 2.3] months; P = 0.01). In multivariate analysis, high CD4 TILs were an independent prognostic factor for disease-free survival (hazard ratio, 0.49; 95% confidence interval, 0.30-0.81; P = 0.005) and OS (hazard ratio, 0.54; 95% confidence interval, 0.33-0.89; P = 0.02). CONCLUSIONS High level of CD4 lymphocytes is associated with tumor differentiation and lower recurrence and is an independent prognostic factor for survival in PDAC patients treated with neoadjuvant therapy.
Collapse
Affiliation(s)
- Reza Nejati
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer B. Goldstein
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Daniel M. Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hua Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nazila Hejazi
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Matthew H. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffrey E. Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason B. Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jorge Blando
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ignacio I. Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gauri R. Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
20
|
Bracci L, Sistigu A, Proietti E, Moschella F. The added value of type I interferons to cytotoxic treatments of cancer. Cytokine Growth Factor Rev 2017; 36:89-97. [PMID: 28693974 DOI: 10.1016/j.cytogfr.2017.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/16/2017] [Indexed: 12/17/2022]
Abstract
Type I interferons (IFNs) exert anti-proliferative, antiviral and immunomodulatory activities. They are also involved in cell differentiation and anti-tumor defense processes. A growing body of literature indicates that the success of conventional chemotherapeutics, epigenetic drugs, targeted anticancer agents and radiotherapy (RT) relies, at least in part, on the induction of type I IFN signaling in malignant cells, tumor-infiltrating antigen presenting cells or other immune cells within lymphoid organs or blood. The mechanisms underlying type I IFN induction and the clinical consequences of these observations are only beginning to be elucidated. In the present manuscript, we reviewed the recent advances in the field and provided our personal view on the role of type I IFNs induced in the context of cytotoxic anticancer treatments and on its possible exploitation as a complement in cancer therapy.
Collapse
Affiliation(s)
- Laura Bracci
- Unit of Tumor Immunology, Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Antonella Sistigu
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy; Department of General Pathology and Physiopathology, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Enrico Proietti
- Unit of Tumor Immunology, Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Federica Moschella
- Unit of Tumor Immunology, Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
21
|
Law AMK, Lim E, Ormandy CJ, Gallego-Ortega D. The innate and adaptive infiltrating immune systems as targets for breast cancer immunotherapy. Endocr Relat Cancer 2017; 24:R123-R144. [PMID: 28193698 PMCID: PMC5425956 DOI: 10.1530/erc-16-0404] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022]
Abstract
A cancer cell-centric view has long dominated the field of cancer biology. Research efforts have focussed on aberrant cancer cell signalling pathways and on changes to cancer cell DNA. Mounting evidence demonstrates that many cancer-associated cell types within the tumour stroma co-evolve and support tumour growth and development, greatly modifying cancer cell behaviour, facilitating invasion and metastasis and controlling dormancy and sensitivity to drug therapy. Thus, these stromal cells represent potential targets for cancer therapy. Among these cell types, immune cells have emerged as a promising target for therapy. The adaptive and the innate immune system play an important role in normal mammary development and breast cancer. The number of infiltrating adaptive immune system cells with tumour-rejecting capacity, primarily, T lymphocytes, is lower in breast cancer compared with other cancer types, but infiltration occurs in a large proportion of cases. There is strong evidence demonstrating the importance of the immunosuppressive role of the innate immune system during breast cancer progression. A consideration of components of both the innate and the adaptive immune system is essential for the design and development of immunotherapies in breast cancer. In this review, we focus on the importance of immunosuppressive myeloid-derived suppressor cells (MDSCs) as potential targets for breast cancer therapy.
Collapse
Affiliation(s)
- Andrew M K Law
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Elgene Lim
- Connie Johnson Breast Cancer Research LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Christopher J Ormandy
- Cancer Biology LaboratoryThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - David Gallego-Ortega
- Tumour Development GroupThe Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
- St. Vincent's Clinical SchoolFaculty of Medicine, University of New South Wales Australia, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Chacon JA, Schutsky K, Powell DJ. The Impact of Chemotherapy, Radiation and Epigenetic Modifiers in Cancer Cell Expression of Immune Inhibitory and Stimulatory Molecules and Anti-Tumor Efficacy. Vaccines (Basel) 2016; 4:E43. [PMID: 27854240 PMCID: PMC5192363 DOI: 10.3390/vaccines4040043] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/17/2016] [Accepted: 11/01/2016] [Indexed: 12/19/2022] Open
Abstract
Genomic destabilizers, such as radiation and chemotherapy, and epigenetic modifiers are used for the treatment of cancer due to their apoptotic effects on the aberrant cells. However, these therapies may also induce widespread changes within the immune system and cancer cells, which may enable tumors to avoid immune surveillance and escape from host anti-tumor immunity. Genomic destabilizers can induce immunogenic death of tumor cells, but also induce upregulation of immune inhibitory ligands on drug-resistant cells, resulting in tumor progression. While administration of immunomodulatory antibodies that block the interactions between inhibitory receptors on immune cells and their ligands on tumor cells can mediate cancer regression in a subset of treated patients, it is crucial to understand how genomic destabilizers alter the immune system and malignant cells, including which inhibitory molecules, receptors and/or ligands are upregulated in response to genotoxic stress. Knowledge gained in this area will aid in the rational design of trials that combine genomic destabilizers, epigenetic modifiers and immunotherapeutic agents that may be synergized to improve clinical responses and prevent tumor escape from the immune system. Our review article describes the impact genomic destabilizers, such as radiation and chemotherapy, and epigenetic modifiers have on anti-tumor immunity and the tumor microenvironment. Although genomic destabilizers cause DNA damage on cancer cells, these therapies can also have diverse effects on the immune system, promote immunogenic cell death or survival and alter the cancer cell expression of immune inhibitor molecules.
Collapse
Affiliation(s)
- Jessica Ann Chacon
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Keith Schutsky
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Daniel J Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Salem ML, Nassef M, Abdel Salam SGR, Zidan A, Mahmoud MH, Badr G, Rubinstein M, Cole D. Effect of administration timing of postchemotherapy granulocyte colony-stimulating factor on host-immune cell recovery and CD8 + T-cell response. J Immunotoxicol 2016; 13:784-792. [PMID: 27417188 PMCID: PMC5669798 DOI: 10.1080/1547691x.2016.1194917] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF), a hematopoietic growth factor, is a standard supportive therapy given during cancer treatment. It induces acceleration in neutrophil recovery through stimulation of mobilization of hematopoietic progenitors. Given that the latter is also induced by chemotherapy itself, the timing of administration of G-CSF postchemotherapy might impact the resultant overall effects. The present study aimed to determine the optimal timing of G-CSF postchemotherapy to exert its optimal effects on the immune cell recovery and its impact on antigen-specific CD8+ T-cell response. B6 mice were treated once with cyclophosphamide (4 mg/mouse; CTX) and then daily with G-CSF (5 g/mouse) from Days 1-5, 2-5 or 5-9 post-CTX treatment. The total numbers of various immune cell types were analyzed on Days 7, 9 and 12 post-CTX treatment. To evaluate effects on CD8+ T-cell response, a pmel-1 transgenic mouse model was used in combination with prime boost peptide vaccination therapy. The total number of white blood cells (WBC), neutrophils, monocytes, lymphocytes, granulocytes and dendritic cells (DC) were significantly increased after G-CSF treatment in particular when G-CSF was administered from Days 2-5 post-CTX treatment. Application of this timing of G-CSF and CTX treatment after adoptive transfer of T-cells followed by prime-boost vaccination with antigenic peptide did not block the expansion of the donor pmel-1 CD8+ T-cells. In conclusion, adjusting the timing of treatment with G-CSF postchemotherapy can optimize its promoting effects on recovery of myeloid cells without altering the associated antigen-specific immunity.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Immunology and Biotechnology Division, Zoology Department, Tanta University, Tanta, Egypt
- Center of Excellence in Cancer Research, Tanta University
| | - Mohamed Nassef
- Immunology and Biotechnology Division, Zoology Department, Tanta University, Tanta, Egypt
| | - Soha G. R. Abdel Salam
- Immunology and Biotechnology Division, Zoology Department, Tanta University, Tanta, Egypt
| | | | - Mohamed H. Mahmoud
- Deanship of Scientific Research, King Saud University, Riyadh, Saudi Arabia
- Food Science and Nutrition Department, National Research Center, Dokki, Cairo, Egypt
| | - Gamal Badr
- Laboratory of Immunology and Molecular Biology, Zoology Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Mark Rubinstein
- Surgery Department and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - David Cole
- Surgery Department and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
24
|
Rini BI, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, Weikert S, Bracarda S, Crabb SJ, Bedke J, Ludwig J, Maurer D, Mendrzyk R, Wagner C, Mahr A, Fritsche J, Weinschenk T, Walter S, Kirner A, Singh-Jasuja H, Reinhardt C, Eisen T. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol 2016; 17:1599-1611. [DOI: 10.1016/s1470-2045(16)30408-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/01/2016] [Accepted: 08/10/2016] [Indexed: 10/20/2022]
|
25
|
Ahlmann M, Hempel G. The effect of cyclophosphamide on the immune system: implications for clinical cancer therapy. Cancer Chemother Pharmacol 2016; 78:661-71. [PMID: 27646791 DOI: 10.1007/s00280-016-3152-1] [Citation(s) in RCA: 287] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/02/2016] [Indexed: 12/17/2022]
Abstract
Cyclophosphamide is an alkylating agent belonging to the group of oxazaphosporines. As cyclophosphamide is in clinical use for more than 40 years, there is a lot of experience using this drug for the treatment of cancer and as an immunosuppressive agent for the treatment of autoimmune and immune-mediated diseases. Besides antimitotic and antireplicative effects, cyclophosphamide has immunosuppressive as well as immunomodulatory properties. Cyclophosphamide shows selectivity for T cells and is therefore now frequently used in tumour vaccination protocols and to control post-transplant allo-reactivity in haplo-identical unmanipulated bone marrow after transplantation. The schedule of administration is of special importance for the immunological effect: while cyclophosphamide can be used in high-dose therapy for the complete eradication of haematopoietic cells, lower doses of cyclophosphamide are relatively selective for T cells. Of special interest is the fact that a single administration of low-dose cyclophosphamide is able to selectively suppress regulatory T cells (Tregs). This effect can be used to counteract immunosuppression in cancer. However, cyclophosphamide can also increase the number of myeloid-derived suppressor cells. Combination of cyclophosphamide with other immunomodulatory agents could be a promising approach to treat different forms of advanced cancer.
Collapse
Affiliation(s)
- Martina Ahlmann
- Pädiatrische Hämatologie und Onkologie, Klinik für Kinder- und Jugendmedizin, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, A1, Münster, Germany
| | - Georg Hempel
- PharmaCampus, Klinische Pharmazie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
26
|
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol 2016; 27:1482-92. [PMID: 27069014 DOI: 10.1093/annonc/mdw168] [Citation(s) in RCA: 812] [Impact Index Per Article: 90.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/06/2016] [Indexed: 12/23/2022] Open
Abstract
The tumor microenvironment (TME) is an integral part of cancer. Recognition of the essential nature of the TME in cancer evolution has led to a shift from a tumor cell-centered view of cancer development to the concept of a complex tumor ecosystem that supports tumor growth and metastatic dissemination. Accordingly, novel targets within the TME have been uncovered that can help direct and improve the actions of various cancer therapies, notably immunotherapies that work by potentiating host antitumor immune responses. Here, we review the composition of the TME, how this attenuates immunosurveillance, and discuss existing and potential strategies aimed at targeting cellular and molecular TME components.
Collapse
Affiliation(s)
- J M Pitt
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre Gustave Roussy Cancer Campus, Villejuif Cedex
| | - A Marabelle
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif INSERM Unit U981, Villejuif
| | - A Eggermont
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif
| | - J-C Soria
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre INSERM Unit U981, Villejuif Drug Development Department (DITEP), Villejuif
| | - G Kroemer
- INSERM U848, Villejuif Metabolomics Platform, GRCC, Villejuif Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM U 1138, Paris Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris Université Paris Descartes, Sorbonne Paris Cité, Paris Université Pierre et Marie Curie, Paris, France Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - L Zitvogel
- Institut de Cancérologie Gustave Roussy Cancer Campus (GRCC), Villejuif INSERM Unit U1015, Villejuif Faculté de Médecine, Université Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre INSERM Unit U932, Institut Curie, Paris Cedex 05 Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, Villejuif, France
| |
Collapse
|
27
|
Noguchi M, Moriya F, Koga N, Matsueda S, Sasada T, Yamada A, Kakuma T, Itoh K. A randomized phase II clinical trial of personalized peptide vaccination with metronomic low-dose cyclophosphamide in patients with metastatic castration-resistant prostate cancer. Cancer Immunol Immunother 2016; 65:151-60. [PMID: 26728480 PMCID: PMC11028889 DOI: 10.1007/s00262-015-1781-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/11/2015] [Indexed: 01/21/2023]
Abstract
This study investigated the effect of metronomic cyclophosphamide (CPA) in combination with personalized peptide vaccination (PPV) on regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC), and whether it could improve the antitumor effect of PPV. Seventy patients with metastatic castration-resistant prostate cancer were randomly assigned (1:1) to receive PPV plus oral low-dose CPA (50 mg/day), or PPV alone. PPV treatment used a maximum of four peptides chosen from 31 pooled peptides according to human leukocyte antigen types and antigen-specific humoral immune responses before PPV, for 8 subcutaneous weekly injections. Peptide-specific cytotoxic T lymphocyte (CTL) and immunoglobulin G responses were measured before and after PPV. The incidence of grade 3 or 4 hematologic adverse events was higher in the PPV plus CPA arm than in the PPV alone arm. Decrease in Treg and increase in MDSC were more pronounced in PPV plus CPA treatment than in PPV alone (p = 0.036 and p = 0.048, respectively). There was no correlation between the changes in Treg or MDSC and CTL response. There was no difference in positive immune responses between the two arms, although overall survival in patients with positive immune responses was longer than in those with negative immune responses (p = 0.001). Significant differences in neither progression-free survival nor overall survival were observed between the two arms. Low-dose CPA showed no change in the antitumor effect of PPV, possibly due to the simultaneous decrease in Treg and increase in MDSC, in patients under PPV.
Collapse
MESH Headings
- Administration, Metronomic
- Aged
- Aged, 80 and over
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/immunology
- Combined Modality Therapy
- Cyclophosphamide/administration & dosage
- Humans
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Metastasis
- Peptides/administration & dosage
- Peptides/chemistry
- Peptides/immunology
- Precision Medicine/methods
- Prostatic Neoplasms, Castration-Resistant/immunology
- Prostatic Neoplasms, Castration-Resistant/mortality
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/therapy
- Survival Analysis
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.
- Department of Urology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan.
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Satoko Matsueda
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuro Sasada
- Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Akira Yamada
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuyuki Kakuma
- Bio-statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
28
|
Kirner A, Mayer-Mokler A, Reinhardt C. IMA901: a multi-peptide cancer vaccine for treatment of renal cell cancer. Hum Vaccin Immunother 2015; 10:3179-89. [PMID: 25625928 DOI: 10.4161/21645515.2014.983857] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Despite a major improvement in the treatment of advanced kidney cancer by the recent introduction of targeted agents such as multi-kinase inhibitors, long-term benefits are still limited and a significant unmet medical need remains for this disease. Cancer immunotherapy has shown its potential by the induction of long-lasting responses in a small subset of patients, however, the unspecific immune interventions with (high dose) cytokines used so far are associated with significant side effects. Specific cancer immunotherapy may circumvent these problems by attacking tumor cells while sparing normal tissue with the use of multi-peptide vaccination being one of the most promising strategies. We here summarize the clinical and translational data from phase I and II trials investigating IMA901. Significant associations of clinical benefit with detectable T cell responses against the IMA901 peptides and encouraging survival data in treated patients has prompted the start of a randomized, controlled phase III trial in 1st line advanced RCC with survival results expected toward the end of 2015. Potential combination strategies with the recently discovered so-called checkpoint inhibitors are also discussed.
Collapse
Key Words
- 5-FU, 5 fluorouracil
- AE, Adverse event
- CTL, Cytotoxic T-lymphocyte
- CY, Cyclophosphamide
- Cancer vaccine
- DC, Dendritic cell
- DCR, Disease control rate
- ECG, Electrocardiogram
- ELISpot, Enzyme-linked immunospot assay
- FDA, Food and Drug Administration
- GM-CSF
- HBV, Hepatitis B virus
- HLA, Human leukocyte antigen
- IFN, Interferon
- IL, Interleukin
- IMA901
- MDSC, Myeloid-derived suppressor cells
- MHC, Major histocompatibility complex
- MSKCC, Memorial Sloan Kettering Cancer Center
- NCI-CTC, National Cancer Institute-Common Toxicity Criteria
- OS, Overall survival
- PD, Progressive disease
- PFS, Progression-free survival
- PK, Pharmacokinetic
- PR, Partial response
- RCC, Renal cell carcinoma
- RECIST, Response Evaluation Criteria in Solid Tumors
- SAE, Serious adverse event
- SD, Stable disease
- TKI, Tyrosine-kinase inhibitors
- TNF, Tumor necrosis factor
- TUMAP, Tumor-associated peptides
- Tregs, Regulatory T-cells
- VEGF, Vascular endothelial growth factor
- ccRCC, Clear cell renal cell carcinoma
- checkpoint inhibitor
- cyclophosphamide
- i.d., intradermal
- immunotherapy
- intradermally
- kidney cancer
- mRNA, Messenger ribonucleic acid
- mTOR, Mammalian target of rapamycin
- mg, Milligram
- n, Number
- renal cell carcinoma
- s.c., subcutaneous, subcutaneously
- tumor-associated peptides
- vaccination
- μg, Microgram
Collapse
|
29
|
Gershan JA, Barr KM, Weber JJ, Jing W, Johnson BD. Immune modulating effects of cyclophosphamide and treatment with tumor lysate/CpG synergize to eliminate murine neuroblastoma. J Immunother Cancer 2015; 3:24. [PMID: 26082836 PMCID: PMC4469315 DOI: 10.1186/s40425-015-0071-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 05/15/2015] [Indexed: 12/22/2022] Open
Abstract
Background Neuroblastoma is a pediatric cancer of neural crest origin. Despite aggressive treatment, mortality remains at 40 % for patients with high-risk disseminated disease, underscoring the need to test new combinations of therapies. In murine tumor models, our laboratory previously showed that T cell-mediated anti-tumor immune responses improve in the context of lymphopenia. The goal of this study was to incorporate lymphodepletion into an effective immune therapy that can be easily translated into neuroblastoma standard of care. Based on the lymphodepleting effects of cyclophosphamide, we hypothesized that cyclophosphamide would synergize with the TLR9 agonist, CpG oligodeoxynucleotide (ODN), to produce a T cell-mediated anti-neuroblastoma effect. Methods To test this hypothesis, we used the AgN2a aggressive murine model of neuroblastoma. Mice bearing subcutaneous tumors were treated with cyclophosphamide followed by treatment with tumor cell lysate mixed with CpG ODN injected at the tumor site. Results Subcutaneous neuroblastoma regressed only in mice that were treated with 100 mg/kg cyclophosphamide prior to receiving treatments of tumor lysate mixed with CpG ODN. The anti-neuroblastoma response was T cell-mediated. Synergy between cyclophosphamide and the tumor lysate/CpG ODN treatment influenced the production of anti-tumor CD8 T cell effectors, and dendritic cell homeostasis. For clinical consideration, an allogeneic tumor lysate was used effectively with this protocol to eliminate AgN2a tumor in vivo. Conclusion Synergistic immune modulating effects of cyclophosphamide and a treatment containing tumor cell lysate and CpG ODN provide T cell-mediated anti-tumor activity against murine neuroblastoma. Electronic supplementary material The online version of this article (doi:10.1186/s40425-015-0071-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jill A Gershan
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | | | - James J Weber
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Weiqing Jing
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Bryon D Johnson
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| |
Collapse
|
30
|
Zavala VA, Kalergis AM. New clinical advances in immunotherapy for the treatment of solid tumours. Immunology 2015; 145:182-201. [PMID: 25826229 PMCID: PMC4427384 DOI: 10.1111/imm.12459] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/08/2015] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Advances in understanding the mechanisms of cancer cells for evading the immune system surveillance, including how the immune system modulates the phenotype of tumours, have allowed the development of new therapies that benefit from this complex cellular network to specifically target and destroy cancer cells. Immunotherapy researchers have mainly focused on the discovery of tumour antigens that could confer specificity to immune cells to detect and destroy cancer cells, as well as on the mechanisms leading to an improved activation of effector immune cells. The Food and Drug Administration approval in 2010 of ipilumumab for melanoma treatment and of pembrolizumab in 2014, monoclonal antibodies against T-lymphocyte-associated antigen 4 and programmed cell death 1, respectively, are encouraging examples of how research in this area can successfully translate into clinical use with promising results. Currently, several ongoing clinical trials are in progress testing new anti-cancer therapies based on the enhancement of immune cell activity against tumour antigens. Here we discuss the general concepts related to immunotherapy and the recent application to the treatment of cancer with positive results that support their consideration of clinical application to patients.
Collapse
Affiliation(s)
- Valentina A Zavala
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiago, Chile
- Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de ChileSantiago, Chile
- INSERM U1064Nantes, France
| |
Collapse
|
31
|
Combination effect of regulatory T-cell depletion and ionizing radiation in mouse models of lung and colon cancer. Int J Radiat Oncol Biol Phys 2015; 92:390-8. [PMID: 25754628 DOI: 10.1016/j.ijrobp.2015.01.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 12/24/2014] [Accepted: 01/12/2015] [Indexed: 11/23/2022]
Abstract
PURPOSE To investigate the potential of low-dose cyclophosphamide (LD-CTX) and anti-CD25 antibody to prevent activation of regulatory T cells (Tregs) during radiation therapy. METHODS AND MATERIALS We used LD-CTX and anti-CD25 monoclonal antibody as a means to inhibit Tregs and improve the therapeutic effect of radiation in a mouse model of lung and colon cancer. Mice were irradiated on the tumor mass of the right leg and treated with LD-CTX and anti-CD25 antibody once per week for 3 weeks. RESULTS Combined treatment of LD-CTX or anti-CD25 antibody with radiation significantly decreased Tregs in the spleen and tumor compared with control and irradiation only in both lung and colon cancer. Combinatorial treatments resulted in a significant increase in the effector T cells, longer survival rate, and suppressed irradiated and distal nonirradiated tumor growth. Specifically, the combinatorial treatment of LD-CTX with radiation resulted in outstanding regression of local and distant tumors in colon cancer, and almost all mice in this group survived until the end of the study. CONCLUSIONS Our results suggest that Treg depletion strategies may enhance radiation-mediated antitumor immunity and further improve outcomes after radiation therapy.
Collapse
|
32
|
Weir GM, Hrytsenko O, Stanford MM, Berinstein NL, Karkada M, Liwski RS, Mansour M. Metronomic cyclophosphamide enhances HPV16E7 peptide vaccine induced antigen-specific and cytotoxic T-cell mediated antitumor immune response. Oncoimmunology 2014; 3:e953407. [PMID: 25960932 PMCID: PMC4368141 DOI: 10.4161/21624011.2014.953407] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 06/24/2014] [Indexed: 12/22/2022] Open
Abstract
In clinical trials, metronomic cyclophosphamide (CPA) is increasingly being combined with vaccines to reduce tumor-induced immune suppression. Previous strategies to modulate the immune system during vaccination have involved continuous administration of low dose chemotherapy, studies that have posed unique considerations for clinical trial design. Here, we evaluated metronomic CPA in combination with a peptide vaccine targeting HPV16E7 in an HPV16-induced tumor model, focusing on the cytotoxic T-cell response and timing of low dose metronomic CPA (mCPA) treatment relative to vaccination. Mice bearing C3 tumors were given metronomic CPA on alternating weeks in combination with immunization with a DepoVax vaccine containing HPV16E749-57 peptide antigen every 3 weeks. Only the combination therapy provided significant long-term control of tumor growth. The efficacy of the vaccine was uncompromised if given at the beginning or end of a cycle of metronomic CPA. Metronomic CPA had a pronounced lymphodepletive effect on the vaccine draining lymph node, yet did not reduce the development of antigen-specific CD8+ T cells induced by vaccination. This enrichment correlated with increased cytotoxic activity in the spleen and increased expression of cytotoxic gene signatures in the tumor. Immunity could be passively transferred through CD8+ T cells isolated from tumor-bearing mice treated with the combinatorial treatment regimen. A comprehensive survey of splenocytes indicated that metronomic CPA, in the absence of vaccination, induced transient lymphodepletion marked by a selective expansion of myeloid-derived suppressor cells. These results provide important insights into the multiple mechanisms of metronomic CPA induced immune modulation in the context of a peptide cancer vaccine that may be translated into more effective clinical trial designs.
Collapse
Key Words
- CPA, cyclophosphamide
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte-associated protein 4
- DPX, DepoVax
- HPV, human papilloma virus
- HPV16
- IFNγ, interferon γ
- MDSC, myeloid-derived suppressor cells
- PD-1/PDCD1, programmed cell death 1
- PO, per os (oral)
- Treg, regulatory T cell
- cancer
- checkpoint inhibitors
- mCPA, metronomic low dose CPA
- metronomic cyclophosphamide
- sbCPA, single bolus low dose CPA
- translational
- vaccine
Collapse
Affiliation(s)
- Genevieve M Weir
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | - Olga Hrytsenko
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Biology; Dalhousie University ; Halifax; Nova Scotia, Cananda
| | - Marianne M Stanford
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | | | - Mohan Karkada
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | - Robert S Liwski
- Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada ; Division of Hematopathology; Queen Elizabeth II Health Sciences Centre ; Nova Scotia, Canada
| | - Marc Mansour
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada
| |
Collapse
|
33
|
Rijavec E, Genova C, Alama A, Barletta G, Sini C, Pronzato P, Coco S, Dal Bello MG, Savarino G, Truini A, Boccardo F, Grossi F. Role of immunotherapy in the treatment of advanced non-small-cell lung cancer. Future Oncol 2014; 10:79-90. [PMID: 24328411 DOI: 10.2217/fon.13.145] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
After several decades of modest results with nonspecific immune stimulants, immunotherapy has become an exciting approach in the treatment of cancer. Although non-small-cell lung cancer has not been considered an immunogenic disease for very long, a better understanding of tumor immunology and the identification of new targets have led to the development of many clinical trials of immune-based therapies for this neoplasm. Promising results from many clinical trials suggest that immunotherapy could be an effective strategy in the management of advanced non-small-cell lung cancer. Further studies are required to help clinicians in the selection of patients who are more likely to benefit from immunotherapy strategies by the identification of biomarkers and to understand when the combination of immunotherapy with other agents should be recommended.
Collapse
Affiliation(s)
- Erika Rijavec
- UOS Tumori Polmonari, IRCCS AOU San Martino IST - Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mittendorf EA, Alatrash G, Xiao H, Clifton GT, Murray JL, Peoples GE. Breast cancer vaccines: ongoing National Cancer Institute-registered clinical trials. Expert Rev Vaccines 2014; 10:755-74. [DOI: 10.1586/erv.11.59] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Chen X, Yang Y, Zhou Q, Weiss JM, Howard OZ, McPherson JM, Wakefield LM, Oppenheim JJ. Effective chemoimmunotherapy with anti-TGFβ antibody and cyclophosphamide in a mouse model of breast cancer. PLoS One 2014; 9:e85398. [PMID: 24416401 PMCID: PMC3887137 DOI: 10.1371/journal.pone.0085398] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/02/2013] [Indexed: 12/26/2022] Open
Abstract
TGFβ is reportedly responsible for accumulation of CD4+Foxp3+ regulatory T cells (Tregs) in tumor. Thus, we treated mouse 4T1 mammary carcinoma with 1D11, a neutralizing anti-TGFβ (1,2,3) antibody. The treatment delayed tumor growth, but unexpectedly increased the proportion of Tregs in tumor. In vitro, 1D11 enhanced while TGFβ potently inhibited the proliferation of Tregs. To enhance the anti-tumor effects, 1D11 was administered with cyclophosphamide which was reported to eliminate intratumoral Tregs. This combination resulted in long term tumor-free survival of up to 80% of mice, and the tumor-free mice were more resistant to re-challenge with tumor. To examine the phenotype of tumor infiltrating immune cells, 4T1-tumor bearing mice were treated with 1D11 and a lower dose of cyclophosphamide. This treatment markedly inhibited tumor growth, and was accompanied by massive infiltration of IFNγ-producing T cells. Furthermore, this combination markedly decreased the number of splenic CD11b+Gr1+ cells, and increased their expression levels of MHC II and CD80. In a spontaneous 4T1 lung metastasis model with resection of primary tumor, this combination therapy markedly increased the survival of mice, indicating it was effective in reducing lethal metastasis burden. Taken together, our data show that anti-TGFβ antibody and cyclophosphamide represents an effective chemoimmunotherapeutic combination.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/pharmacology
- Antineoplastic Agents, Alkylating/pharmacology
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- CD11b Antigen/genetics
- CD11b Antigen/immunology
- Carcinoma/immunology
- Carcinoma/mortality
- Carcinoma/pathology
- Carcinoma/therapy
- Cell Line, Tumor
- Cyclophosphamide/pharmacology
- Drug Synergism
- Drug Therapy, Combination
- Female
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression/immunology
- Humans
- Immunotherapy
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Lymphocyte Count
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Survival Analysis
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/immunology
Collapse
Affiliation(s)
- Xin Chen
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, United States of America
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail: (XC); (JJO)
| | - Yuan Yang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Qiong Zhou
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Jonathan M. Weiss
- Laboratory of Experimental Immunology, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - OlaMae Zack Howard
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - John M. McPherson
- Genzyme Corporation, Framingham, Massachusetts, United States of America
| | - Lalage M. Wakefield
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Joost J. Oppenheim
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
- * E-mail: (XC); (JJO)
| |
Collapse
|
36
|
Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ 2014; 21:15-25. [PMID: 23787994 PMCID: PMC3857622 DOI: 10.1038/cdd.2013.67] [Citation(s) in RCA: 673] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Conventional anticancer chemotherapy has been historically thought to act through direct killing of tumor cells. This concept stems from the fact that cytotoxic drugs interfere with DNA synthesis and replication. Accumulating evidence, however, indicates that the antitumor activities of chemotherapy also rely on several off-target effects, especially directed to the host immune system, that cooperate for successful tumor eradication. Chemotherapeutic agents stimulate both the innate and adaptive arms of the immune system through several modalities: (i) by promoting specific rearrangements on dying tumor cells, which render them visible to the immune system; (ii) by influencing the homeostasis of the hematopoietic compartment through transient lymphodepletion followed by rebound replenishment of immune cell pools; (iii) by subverting tumor-induced immunosuppressive mechanisms and (iv) by exerting direct or indirect stimulatory effects on immune effectors. Among the indirect ways of immune cell stimulation, some cytotoxic drugs have been shown to induce an immunogenic type of cell death in tumor cells, resulting in the emission of specific signals that trigger phagocytosis of cell debris and promote the maturation of dendritic cells, ultimately resulting in the induction of potent antitumor responses. Here, we provide an extensive overview of the multiple immune-based mechanisms exploited by the most commonly employed cytotoxic drugs, with the final aim of identifying prerequisites for optimal combination with immunotherapy strategies for the development of more effective treatments against cancer.
Collapse
Affiliation(s)
- L Bracci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - G Schiavoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A Sistigu
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - F Belardelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
37
|
Abstract
Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.
Collapse
Affiliation(s)
- Mohamed L Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
38
|
Dudek AM, Martin S, Garg AD, Agostinis P. Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity. Front Immunol 2013; 4:438. [PMID: 24376443 PMCID: PMC3858649 DOI: 10.3389/fimmu.2013.00438] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/23/2013] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) are the sentinel antigen-presenting cells of the immune system; such that their productive interface with the dying cancer cells is crucial for proper communication of the "non-self" status of cancer cells to the adaptive immune system. Efficiency and the ultimate success of such a communication hinges upon the maturation status of the DCs, attained following their interaction with cancer cells. Immature DCs facilitate tolerance toward cancer cells (observed for many apoptotic inducers) while fully mature DCs can strongly promote anticancer immunity if they secrete the correct combinations of cytokines [observed when DCs interact with cancer cells undergoing immunogenic cell death (ICD)]. However, an intermediate population of DC maturation, called semi-mature DCs exists, which can potentiate either tolerogenicity or pro-tumorigenic responses (as happens in the case of certain chemotherapeutics and agents exerting ambivalent immune reactions). Specific combinations of DC phenotypic markers, DC-derived cytokines/chemokines, dying cancer cell-derived danger signals, and other less characterized entities (e.g., exosomes) can define the nature and evolution of the DC maturation state. In the present review, we discuss these different maturation states of DCs, how they might be attained and which anticancer agents or cell death modalities (e.g., tolerogenic cell death vs. ICD) may regulate these states.
Collapse
Affiliation(s)
- Aleksandra M Dudek
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Shaun Martin
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Abhishek D Garg
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| |
Collapse
|
39
|
Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of immunogenic cancer cell death. Cytokine Growth Factor Rev 2013; 24:319-33. [DOI: 10.1016/j.cytogfr.2013.01.005] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/09/2013] [Indexed: 02/07/2023]
|
40
|
Arens R, van Hall T, van der Burg SH, Ossendorp F, Melief CJM. Prospects of combinatorial synthetic peptide vaccine-based immunotherapy against cancer. Semin Immunol 2013; 25:182-90. [PMID: 23706598 DOI: 10.1016/j.smim.2013.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 04/10/2013] [Accepted: 04/19/2013] [Indexed: 01/15/2023]
Abstract
The insight that the immune system is involved in tumor resistance is gaining momentum and this has led to the development of immunotherapeutic strategies aiming at enhancement of immune-mediated tumor destruction. Although some of these strategies have moderate clinical benefit, most stand-alone therapies fail to significantly affect progressive disease and survival or do so only in a minority of patients. Research on the mechanisms underlying the generation of immune responses against tumors and the immune evasion by tumors has emphasized that various mechanisms simultaneously prevent effective immunity against cancer including inefficient presentation of tumor antigens by dendritic cells and induction of negative immune regulation by regulatory T-cells (Tregs) and myeloid derived suppressor cells (MDSCs). Thus the design of therapies that simultaneously improve effective tumor immunity and counteract immune evasion by tumors seems most desirable for clinical efficacy. As it is unlikely that a single immunotherapeutic strategy addresses all necessary requirements, combinatorial strategies that act synergistically need to be developed. Here we discuss the current knowledge and prospects of treatment with synthetic peptide vaccines that stimulate tumor-specific T-cell responses combined with adjuvants, immune modulating antibodies, cytokines and chemotherapy. We conclude that combinatorial approaches have the best potency to accomplish the most significant tumor destruction but further research is required to optimize such approaches.
Collapse
Affiliation(s)
- Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
41
|
Volk A, Hartmann S, Muik A, Geiss Y, Königs C, Dietrich U, von Laer D, Kimpel J. Comparison of three humanized mouse models for adoptive T cell transfer. J Gene Med 2013; 14:540-8. [PMID: 22847974 DOI: 10.1002/jgm.2652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Humanized mouse models for adoptive T cell transfer are important for preclinical efficacy and toxicity studies. However, common xenograft models using immunodeficient mice have so far failed to efficiently support the homing of human T cells to secondary lymphoid tissues. METHODS We established a new mouse model for the adoptive transfer of genetically-modified (gm) T cells using conditioned BALB/c mice. Conditioning involved cyclophosphamide injections, lethal irradiation and radioprotection with bone marrow from immunodeficient mice. We compared repopulation kinetics and the quality of grafts in these modified Trimera (mT3) mice with immunodeficient BALB/c Rag2(-/-) interleukin (IL)2 receptor gamma (rg) knockout (DKO) and NOD/LtSz-scid IL2rg(-/-) (NSG) recipient mouse strains. RESULTS DKO mice showed only marginal engraftment until onset of graft-versus-host disease, whereas mT3 and NSG were repopulated with comparable kinetics. However, T cell repertoire and human cytokine profiles suggest a xenoreactivity-driven gm T cell expansion in mT3 mice, whereas NSG mice were characterized by an initial homeostatic proliferation. Morphological analysis revealed high levels of human gm T cell infiltration in the spleen and liver of all three mouse strains. However, mT3 mice provided the strongest homing of human gm T cells to mucosal sites. Additionally, mT3 mice were the only model with macroscopically visible superficial inguinal lymph nodes. These lymph nodes strongly supported the homing of gm T cells. CONCLUSIONS In the present study, we give proof-of-concept that wild-type mice can accept gm T cell grafts while providing secondary lymphoid structures. Despite limitations, mT3 mice are a valid alternative for applications that specifically rely on improved secondary lymphoid structures.
Collapse
Affiliation(s)
- Andreas Volk
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bergamaschi C, Bear J, Rosati M, Beach RK, Alicea C, Sowder R, Chertova E, Rosenberg SA, Felber BK, Pavlakis GN. Circulating IL-15 exists as heterodimeric complex with soluble IL-15Rα in human and mouse serum. Blood 2012; 120:e1-8. [PMID: 22496150 PMCID: PMC3390963 DOI: 10.1182/blood-2011-10-384362] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 03/24/2012] [Indexed: 01/24/2023] Open
Abstract
IL-15 is an important cytokine for the function of the immune system, but the form(s) of IL-15 produced in the human body are not fully characterized. Coexpression of the single-chain IL-15 and the IL-15 receptor alpha (IL-15Rα) in the same cell allows for efficient production, surface display, and eventual cleavage and secretion of the bioactive IL-15/IL-15Rα heterodimer in vivo, whereas the single-chain IL-15 is poorly secreted and unstable. This observation led to the hypothesis that IL-15 is produced and secreted only as a heterodimer with IL-15Rα. We purified human IL-15/IL-15Rα complexes from overproducing human cell lines and developed an ELISA specifically measuring the heterodimeric form of IL-15. Analysis of sera from melanoma patients after lymphodepletion revealed the presence of circulating IL-15/IL-15Rα complexes in amounts similar to the total IL-15 quantified by a commercial IL-15 ELISA that detects both the single-chain and the heterodimeric forms of the cytokine. Therefore, in lymphodepleted cancer patients, the serum IL-15 is exclusively present in its heterodimeric form. Analysis of the form of IL-15 present in either normal or lymphodepleted mice agrees with the human data. These results have important implications for development of assays and materials for clinical applications of IL-15.
Collapse
Affiliation(s)
- Cristina Bergamaschi
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Diaz-Montero CM, Wang Y, Shao L, Feng W, Zidan AA, Pazoles CJ, Montero AJ, Zhou D. The glutathione disulfide mimetic NOV-002 inhibits cyclophosphamide-induced hematopoietic and immune suppression by reducing oxidative stress. Free Radic Biol Med 2012; 52:1560-8. [PMID: 22343421 PMCID: PMC3341494 DOI: 10.1016/j.freeradbiomed.2012.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 01/20/2012] [Accepted: 02/04/2012] [Indexed: 12/22/2022]
Abstract
The oxidized glutathione mimetic NOV-002 is a unique anti-tumor agent that not only has the ability to inhibit tumor cell proliferation, survival, and invasion, but in some settings can also ameliorate cytotoxic chemotherapy-induced hematopoietic and immune suppression. However, the mechanisms by which NOV-002 protects the hematopoietic and immune systems against the cytotoxic effects of chemotherapy are not known. Therefore, in this study we investigated the mechanisms of action of NOV-002 using a mouse model in which hematopoietic and immune suppression was induced by cyclophosphamide (CTX) treatment. We found that NOV-002 treatment in a clinically comparable dose regimen attenuated CTX-induced reduction in bone marrow hematopoietic stem and progenitor cells (HSPCs) and reversed the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), which led to a significant improvement in hematopoietic and immune functions. These effects of NOV-002 may be attributable to its ability to modulate cellular redox. This suggestion is supported by the finding that NOV-002 treatment upregulated the expression of superoxide dismutase 3 and glutathione peroxidase 2 in HSPCs, inhibited CTX-induced increases in reactive oxygen species production in HSPCs and MDSCs, and attenuated CTX-induced reduction of the ratio of reduced glutathione to oxidized glutathione in splenocytes. These findings provide a better understanding of the mechanisms whereby NOV-002 modulates chemotherapy-induced myelosuppression and immune dysfunction and a stronger rationale for clinical utilization of NOV-002 to reduce chemotherapy-induced hematopoietic and immune suppression.
Collapse
Affiliation(s)
| | - Yong Wang
- Department of Pathology, Medical University of South Carolina, Charleston, SC 29425
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Wei Feng
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Abdel-Aziz Zidan
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | | | - Alberto J. Montero
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Daohong Zhou
- Department of Pathology, Medical University of South Carolina, Charleston, SC 29425
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| |
Collapse
|
44
|
Arevalo-Martin A, Molina-Holgado E, Guaza C. A CB₁/CB₂ receptor agonist, WIN 55,212-2, exerts its therapeutic effect in a viral autoimmune model of multiple sclerosis by restoring self-tolerance to myelin. Neuropharmacology 2012; 63:385-93. [PMID: 22561283 DOI: 10.1016/j.neuropharm.2012.04.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/19/2012] [Accepted: 04/17/2012] [Indexed: 01/05/2023]
Abstract
Infection of mice with Theiler's murine encephalomyelitis virus (TMEV) leads to the development of TMEV-induced demyelinating disease (TMEV-IDD), an autoimmune, demyelinating and neurodegenerative pathology that serves as a model of multiple sclerosis. Activation of endogenous CB₁/CB₂ cannabinoid receptors inhibits inflammation and improves the clinical status of TMEV-IDD animals. In the present study, mice with established TMEV-IDD were treated with the CB₁/CB₂ receptor agonist WIN 55,212-2 (WIN), which restored self-tolerance to a myelin self-antigen while ameliorating the disease in a long-term manner. Accordingly, disruption of self-tolerance with cyclophosphamide provoked chronic relapse. Furthermore, transfer of splenocytes from WIN-treated TMEV-IDD mice to TMEV-infected mice at disease onset prevented the autoimmune inflammatory response and motor impairment. The therapeutic effect of WIN correlated with a decrease in the activation of CD4⁺CD25⁺Foxp3⁻ T cells and an increase in regulatory CD4⁺CD25⁺Foxp3⁺ T cells in the CNS, along with alterations in the cytokine and chemokine milieu. These findings demonstrate for the first time that the suppression of autoimmune responses to myelin antigens underlies the therapeutic effect of CB₁/CB₂ cannabinoid agonists in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Angel Arevalo-Martin
- Laboratorio de Neuroinflamación, Hospital Nacional de Paraplejicos, SESCAM, Finca la Peraleda s/n., Toledo 45071, Spain.
| | | | | |
Collapse
|
45
|
Hadaschik B, Su Y, Huter E, Ge Y, Hohenfellner M, Beckhove P. Antigen Specific T-Cell Responses Against Tumor Antigens are Controlled by Regulatory T Cells in Patients With Prostate Cancer. J Urol 2012; 187:1458-65. [DOI: 10.1016/j.juro.2011.11.083] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Indexed: 11/28/2022]
Affiliation(s)
- Boris Hadaschik
- Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
| | - Yun Su
- Department of Urology, University Hospital Heidelberg, Heidelberg, Germany
- Department of Urology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Eva Huter
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Yingzi Ge
- Translational Immunology Unit, The German Cancer Research Center, Heidelberg, Germany
| | | | - Philipp Beckhove
- Translational Immunology Unit, The German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
46
|
Rubinstein MP, Cloud CA, Garrett TE, Moore CJ, Schwartz KM, Johnson CB, Craig DH, Salem ML, Paulos CM, Cole DJ. Ex vivo interleukin-12-priming during CD8(+) T cell activation dramatically improves adoptive T cell transfer antitumor efficacy in a lymphodepleted host. J Am Coll Surg 2012; 214:700-7; discussion 707-8. [PMID: 22360982 PMCID: PMC3429131 DOI: 10.1016/j.jamcollsurg.2011.12.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 12/15/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical application of adoptive T cell therapy has been hindered by an inability to generate adequate numbers of nontolerized, functionally active, tumor-specific T cells, which can persist in vivo. In order to address this, we evaluated the impact of interleukin (IL)-12 signaling during tumor-specific CD8(+) T cell priming in terms of persistence and antitumor efficacy using an established B16 melanoma tumor adoptive therapy model. STUDY DESIGN B6 mice were injected subcutaneously with B16 melanoma tumor cells. On day 12 of tumor growth, mice were preconditioned with cyclophosphamide (4mg dose, intraperitoneally), and 1 day later were treated by adoptive transfer of tumor-specific pmel-1 CD8(+) T cells primed ex vivo 3 days earlier with both IL-12 and antigen (hGP100(25-33) peptide) or antigen only. Tumors were measured biweekly, and infused donor T cells were analyzed for persistence, localization to the tumor, phenotype, and effector function. RESULTS Adoptive transfer of tumor-specific CD8(+) T cells primed with IL-12 was significantly more effective in reducing tumor burden in mice preconditioned with cyclophosphamide compared with transfer of T cells primed without IL-12. This enhanced antitumor response was associated with increased frequencies of infused T cells in the periphery and tumor as well as elevated expression of effector molecules including granzyme B and interferon-γ (IFNγ). CONCLUSIONS Our findings demonstrate that ex vivo priming of tumor-specific CD8(+) T cells with IL-12 dramatically improves their in vivo persistence and therapeutic ability on transfer to tumor-bearing mice. These findings can be directly applied as novel clinical trial strategies.
Collapse
Affiliation(s)
- Mark P Rubinstein
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
For a long time, anticancer therapies were believed to work (and hence convey a therapeutic benefit) either by killing cancer cells or by inducing a permanent arrest in their cell cycle (senescence). In both scenarios, the efficacy of anticancer regimens was thought to depend on cancer cell-intrinsic features only. More recently, the importance of the tumor microenvironment (including stromal and immune cells) has been recognized, along with the development of therapies that function by modulating tumor cell-extrinsic pathways. In particular, it has been shown that some chemotherapeutic and radiotherapeutic regimens trigger cancer cell death while stimulating an active immune response against the tumor. Such an immunogenic cell death relies on the coordinated emission of specific signals from dying cancer cells and their perception by the host immune system. The resulting tumor-specific immune response is critical for the eradication of tumor cells that may survive therapy. In this review, we discuss the molecular mechanisms that underlie the vaccine-like effects of some chemotherapeutic and radiotherapeutic regimens, with particular attention to the signaling pathways and genetic elements that constitute the prerequisites for immunogenic anticancer therapy.
Collapse
|
48
|
Salem ML, Al-Khami AA, El-Nagaar SA, Zidan AAA, Al-Sharkawi IM, Marcela Díaz-Montero C, Cole DJ. Kinetics of rebounding of lymphoid and myeloid cells in mouse peripheral blood, spleen and bone marrow after treatment with cyclophosphamide. Cell Immunol 2012; 276:67-74. [PMID: 22560674 PMCID: PMC3787597 DOI: 10.1016/j.cellimm.2012.03.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 03/06/2012] [Accepted: 03/21/2012] [Indexed: 02/07/2023]
Abstract
Recently, we showed that post cyclophosphamide (CTX) microenvironment benefits the function of transferred T cells. Analysis of the kinetics of cellular recovery after CTX treatment showed that a single 4 mg/mouse CTX treatment decreased the absolute number of leukocytes in the peripheral blood (PBL) at days 3-15, and in the spleen and bone marrow (BM) at days 3-6. The absolute numbers of CD11c(+)CD11b(-) and CD11c(+)CD11b(+) dendritic cells (DCs), CD11b(+) and Ly6G(+) myeloid cells, T and B cells, CD4(+)CD25(+) T regulatory (T(reg)) cells, and NK1.1(+) cells also decreased. The cell numbers returned to control levels during the recovery phase. The absolute numbers of B cells remained low for 3 weeks. The numbers of DCs increased in PBL and spleen at day 9 but returned to control levels at day 15. These data indicate that CTX alters the cellular microenvironment in kinetics that might be precisely targeted to benefit the host.
Collapse
Affiliation(s)
- Mohamed L Salem
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | | | | | | | | | | | | |
Collapse
|
49
|
Chemotherapeutics and radiation stimulate MHC class I expression through elevated interferon-beta signaling in breast cancer cells. PLoS One 2012; 7:e32542. [PMID: 22396773 PMCID: PMC3291570 DOI: 10.1371/journal.pone.0032542] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 01/29/2012] [Indexed: 12/21/2022] Open
Abstract
Low doses of anticancer drugs have been shown to enhance antitumor immune response and increase the efficacy of immunotherapy. The molecular basis for such effects remains elusive, although selective depletion of T regulatory cells has been demonstrated. In the current studies, we demonstrate that topotecan (TPT), a topoisomerase I-targeting drug with a well-defined mechanism of action, stimulates major histocompatibility complex class I (MHC I) expression in breast cancer cells through elevated expression/secretion of interferon-β (IFN-β) and activation of type I IFN signaling. First, we show that TPT treatment elevates the expression of both total and cell-surface MHC I in breast cancer cells. Second, conditioned media from TPT-treated breast cancer ZR-75-1 cells induce elevated expression of cell-surface MHC I in drug-naïve recipient cells, suggesting the involvement of cytokines and/or other secreted molecules. Consistently, TPT-treated cells exhibit elevated expression of multiple cytokines such as IFN-β, TNF-α, IL-6 and IL-8. Third, either knocking down the type I interferon receptor subunit 1 (IFNAR1) or addition of neutralizing antibody against IFN-β results in reduced MHC I expression in TPT-treated cells. Together, these results suggest that TPT induces increased IFN-β autocrine/paracrine signaling through type I IFN receptor, resulting in the elevated MHC I expression in tumor cells. Studies have also demonstrated that other chemotherapeutic agents (e.g. etoposide, cisplatin, paclitaxel and vinblastine) similarly induce increased IFN-β secretion and elevated MHC I expression. In addition, conditioned media from γ-irradiated donor cells are shown to induce IFN-β-dependent MHC I expression in unirradiated recipient cells. In the aggregate, our results suggest that many cancer therapeutics induce elevated tumor antigen presentation through MHC I, which could represent a common mechanism for enhanced antitumor immune response through T cell cytotoxicity during metronomic chemotherapy, as well as increased efficacy of combined chemo- (or radio-)/immuno-therapy.
Collapse
|
50
|
Cytotoxic chemotherapy and CD4+ effector T cells: an emerging alliance for durable antitumor effects. Clin Dev Immunol 2012; 2012:890178. [PMID: 22400040 PMCID: PMC3286900 DOI: 10.1155/2012/890178] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 11/01/2011] [Accepted: 11/05/2011] [Indexed: 12/18/2022]
Abstract
Standard cytotoxic chemotherapy can initially achieve high response rates, but relapses often occur in patients and represent a severe clinical problem. As increasing numbers of chemotherapeutic agents are found to have immunostimulatory effects, there is a growing interest to combine chemotherapy and immunotherapy for synergistic antitumor effects and improved clinical benefits. Findings from recent studies suggest that highly activated, polyfunctional CD4+ effector T cells have tremendous potential in strengthening and sustaining the overall host antitumor immunity in the postchemotherapy window. This review focuses on the latest progresses regarding the impact of chemotherapy on CD4+ T-cell phenotype and function and discusses the prospect of exploiting CD4+ T cells to control tumor progression and prevent relapse after chemotherapy.
Collapse
|