Abstract
The alkylating agent melphalan is used in the treatment of hematological malignancies, especially multiple myeloma. In the past, the usefulness of melphalan has been solely attributed to its cytotoxicity on fast-growing cancerous cells. Although the immunomodulatory effects of melphalan were suggested many years ago, only recently has this aspect of melphalan’s activity begun to be elucidated at the molecular level. Emerging evidence indicates that melphalan can foster an immunogenic microenvironment by inducing immunogenic cell death (ICD) as characterized by membrane translocation of endoplasmic reticulum protein calreticulin (CRT) and by release of chromatin-binding protein high-mobility group box 1 (HMGB1). In addition, the lympho-depletive effect of melphalan can induce the release of pro-inflammatory cytokines and growth factors, deplete regulatory T cells, and create space to facilitate the expansion of infused tumor-reactive T cells. These features suggest that melphalan can be used as a preparative chemotherapy for adoptive T-cell therapy. This notion is supported by our recent work demonstrating that the combination of melphalan and adoptive transfer of tumor-reactive CD4+ T cells can mediate potent antitumor effects in animal models. This review summarizes the recent advances in understanding and utilizing the immunomodulatory effects of melphalan.
Keywords: melphalan, CD4, immunostimulation, adoptive cell therapy
I. INTRODUCTION
Chemotherapy is a major treatment modality for many types of cancer. Chemotherapy can often improve the symptoms of cancer initially, but it is rarely curative. Effective treatment of relapsed/refractory cancer remains a major unmet medical need. Recent advances in cancer immunotherapy have offered unprecedented opportunity to harness the power of the immune system to fight against cancer.1 Interest in combining conventional chemotherapy with immunotherapy to address the problem of cancer recurrence in patients is growing.2,3 Therefore, identifying anticancer drugs with immunopotentiating effects can broaden the usefulness of chemotherapy and achieve durable therapeutic benefits.
In recent years, an increasing number of chemotherapeutic agents have been found to exert immunostimulatory effects.4–8 Among these antineoplastic agents, alkylating agent cyclophosphamide (CTX) represents the most well-studied immunopotentiating anticancer drug. Because of its potent immunostimulatory effects, CTX has been widely used both in preclinical studies and clinical settings to enhance the efficacy of cancer immunotherapies.9–11 Melphalan is another alkylating agent widely used in cancer chemotherapy. Although melphalan belongs to the same class of anticancer drug as CTX, little attention has been given to its potential immunomodulatory effects. Our recent work provides the first evidence that melphalan resembles CTX in multiple aspects of immunostimulatory properties, justifying its novel use as an immunopotentiating anticancer drug in the context of combinatorial chemo-immunotherapy. Herein, we discuss our current knowledge of melphalan’s immunomodulatory effects and its potential usage in adoptive immunotherapy.
II. USE OF MELPHALAN IN CANCER TREATMENT
Melphalan (trade name Alkeran) belongs to the class of nitrogen mustard alkylating agents. Since Gilman and Philips published their initial results of treating neoplasms using mustards,12 many drugs in this class, including cyclophosphamide (CTX), melphalan, chlorambucil, uramustine, ifosfamide, and bendamustine, have been developed and used in chemotherapy for cancer. Melphalan was synthesized in the early 1950s by Bergel and Stock, and it was subsequently shown that mainly the L-form had biological effect on tumor cells.13 Unlike CTX, which is a prodrug that needs to be activated by liver through oxidation of cytochrome P450, melphalan does not require any activation to exert its toxicity. Melphalan enters cells primarily by a neutral amino acid active pathway shared by leucine (and therefore its transport is decreased in the event of high leucine concentration).14 It has a half-life of 75 minutes in vivo and is excreted mostly through urine.15 Melphalan exerts its cytotoxicity by inducing inter-strand cross-links in DNA,16 and it may also induce lesions in RNA, proteins, and lipids.17
Melphalan has been used in the treatment of lymphoma and leukemia and several types of solid tumor, including neuroblastoma, melanoma, sarcoma, and ovarian cancer.18 Melphalan is best known for its use in the treatment of multiple myeloma (MM). Melphalan was first used for multiple myeloma in 1962 as a frontline therapy, significantly extending patients’ lives.19 Since prednisone was also shown to have beneficial effect in MM patients, melphalan/prednisone therapy was introduced in 1969; it resulted in increased response rates and median survival of six months over melphalan alone.20 Thalidomide and its derivative lenalidomide, which are immunomodulatory drugs and potent angiogenesis inhibitors,21,22 and bortezomib, a proteasome inhibitor, have been introduced into the therapeutic regimens and have been shown to increase the overall survival of MM patients over other combinations used so far.23,24 Currently, melphalan is included in regimens like MPB (melphalan-prednisone-bortezomib) for initial treatment of MM,25 or BMPT (bortezomib-melphalan-prednisone-thalidomide) in case of refractory tumor.26 Because of its ablative effect toward bone marrow, melphalan has been used in combination with autologous stem cell transplantation (ASCT). High-dose melphalan (HDM; range: 140–200 mg/m2) plus ASCT currently serves as the standard treatment approach for patients with newly diagnosed, transplant-eligible multiple myeloma.27 Low-dose melphalan (LDM, range: 100–140 mg/m2) in combination with other drugs, such as prednisone and thalidomide, is used for patients not eligible for stem cells transplantation.28,29 Its potent immunosuppressive property is also exploited in allogeneic stem cell transfer to allow graft-versus-tumor effects of the transplanted cells.13
Melphalan therapy induces expression of reactive oxygen species that lead to apoptosis through activation of caspase-9, resulting in activation of caspase-3 and subsequent DNA damages that lead to cell death.30,31 In addition to acting on fast-growing transformed cells, alkylating agents also have detrimental activities toward normal tissues including intestine, liver, kidneys, and lungs. The gastrointestinal effects of melphalan are common among patients, causing symptoms such as vomiting, nausea, and/or diarrhea.32 High-dose melphalan plus ASCT causes severe mucositis, limiting the dose of melphalan to 200 mg/m2.33,34 Melphalan also shows toxicity toward liver, kidneys, and lungs, and these side effects need to be accounted for when used in combination with other drugs having similar effects.35 Without ASCT, high-dose melphalan causes severe myelosuppression, which affects cells of the immune origin including T and B lymphocytes and NK cells.36,37
III. IMMUNOMODULATORY EFFECTS OF MELPHALAN
In recent years, it has been realized that in addition to exerting direct cytotoxicity on fast-growing transformed cells, many chemotherapeutic agents can incite antitumor immune responses, which contribute to the efficacy of chemotherapy.38–40 It has been shown that some anticancer drugs, including CTX, doxorubicin, and oxaliplatin, can induce immunogenic cell death (ICD), characterized by surface exposure of the endoplasmic reticulum protein calreticulin (CRT), secretion of chromatin-binding protein high-mobility group box 1 (HMGB1), and release of ATP.41–44 Translocation of CRT from the endoplasmic reticulum to tumor cell surface causes an “eat-me” signal for phagocytosis by dendritic cells (DCs). HMGB1 released by dying tumor cells can act upon TLR4 on DCs. ATP released by dying tumor cells can trigger purinergic P2RX7 receptors on DCs to activate the NLRP3 inflammasome, thus inducing the production of the proinflammatory cytokine IL-1β. These events can lead to enhanced antigen processing and presentation, which result in the activation of endogenous CD8+ T cells, which in turn contribute to the antitumor efficacy of chemotherapy. In addition, certain chemotherapeutic agents can potentiate antitumor immune responses by mitigating immunosuppression mediated by regulatory T cells (Tregs) or myeloid-derived suppressor cells (MDSCs). Low-dose CTX is capable of depleting cycling CD4+CD25+ Tregs and inhibiting their suppressive activity.45,46 Selective and efficient depletion of Tregs and sparing of effector CD4+ cells is highly important because on top of thymic-derived Treg cells, regulatory T cells can arise from the conversion of non-Treg helper cells, especially in tumor microenvironment.47 These de novo generated Tregs have a suppressive phenotype similar to thymic Tregs, yet they possess an unstable phenotype that depends largely on their context or environment.48 Gemcitabine, 5-fluorouracil, sunitinib, doxorubicin, and docetaxel can reduce MDSCs and enhance the antitumor activities of CD8+ T cells and NK cells.49–53 Furthermore, CTX can induce a surge of growth factors and proinflammatory cytokines/chemokines by removing “cytokine sinks,” resulting in reprogramming of the tumor immune milieu from immunosuppressive to immunostimulatory.43,44,54
CTX is probably the most well-studied anticancer drug that possesses all of the immunostimulatory properties described above. Compared with CTX, less attention has been given to melphalan in terms of its immunomodulatory effects. Nonetheless, it has been known for years that low-dose melphalan (LDM) induces antitumor immune responses in mouse tumor models. In a series of studies, Mokyr’s group showed that LDM (2.5 mg/kg) therapy of mice bearing plasmacytoma MOPC315 tumors was able to induce robust CD8+ T-cell responses that eradicated large tumors.55 These tumor-reactive CD8+ T cells can originate not only from the spleen but also from the thymus. The same group later showed that low-dose melphalan treatment resulted in upregulation of B7.1 not only in tumor cells but also other nontumor cells in the stroma, likely antigen-presenting cells, which may contribute to improved antitumor effects.56
It has been well-established that CTX can exert immunostimulatory effects by promoting the release of pro-inflammatory cytokines (removing “cytokine sinks”), inducing immunogenic cell death, reducing Tregs and MDSCs, and creating space to facilitate T-cell expansion.7,42,45,57,58 Although it is reasonable to assume that melphalan has similar immunomodulatory effects as CTX because they belong to the same class of alkylating agent, supporting clinical and experimental evidence has been lacking until recently. It has been shown that myeloma patients had increased plasma level of immune cytokines, including IL-6, IL-7 and IL-15, following HDM and ASCT, and the presence of IL-7 and IL-15 may contribute to the survival and activation of CD3+ T cells present in the SCT graft.37 Along this line, the recent report by Dudek-Perić et al.39 showed that limb perfusion of melanoma patients with melphalan led to increased release of IL-1β, IL-6, and IL-8 in the locoregional sera of patients. In a prophylactic vaccination mouse model, the authors showed that melphalan administration in vivo could stimulate a CD8+ T-cell-dependent protective antitumor response. Interestingly, although induction of apoptosis in melanoma cells by melphalan in vitro did not elicit cell surface expression of CRT, vaccination effect was potentiated in combination with exogenous CRT.
Our recent work showed for the first time that melphalan is able to induce ICD in tumor cells in vitro and in vivo.59 We showed that melphalan treatment led to expression of CRT on the surface of A20 (B-cell lymphoma) and CT26 (colorectal cancer) cells, accompanied by the release of extracellular HMGB1. Notably, the ability of melphalan to induce CRT translocation may be cell dependent: CRT surface expression was not observed in melanoma cells after melphalan treatment even through the treatment triggered inflammatory cell death.60 In our study, the emergence of immunogenic danger signals correlated with increased uptake of tumor associated antigens by dendritic cells as well as activation of the endogenous CD8+ T cells.57 Our study also demonstrated that melphalan is lympho- and myelo-depletive, causing transient reduction of Tregs and MDSCs. In addition, melphalan induces a proinflammatory cytokine/chemokine milieu, resulting in spikes in the level of cytokines including IFN-γ, IL-22, IL-10, IL-5, IL-18, and IL-27, and chemokines including CCL2, CCL7, CXCL1, and CXCL10. These data provide clear evidence that melphalan resembles CTX in immunomodulatory activities.
IV. THE USE OF MELPHALAN IN CANCER IMMUNOTHERAPY
A. Combination of Melphalan-Based Care and Adoptive T-Cell Therapy for Multiple Myeloma
Commonly, chemotherapy can reduce the symptoms of cancer initially, but refractory cancers develop and become resistant to further treatment.61 This phenomenon suggests that the endogenous antitumor immune responses incited by chemotherapy are insufficient to mediate durable antitumor effects. Therefore, additional therapeutic interventions are needed to confer long-term, curative efficacy. Recent advances in immune checkpoint blockade, therapeutic cancer vaccines, and adoptive T-cell therapy (ACT) have validated immunotherapy as a viable treatment option for patients with cancer.1,62–64 There is increasing interest in combining immunotherapy with conventional cancer therapies to improve patient outcomes. Because chemotherapy is the stand of care for many types of cancer, the concept of combining chemotherapy and immunotherapy for synergistic effect has gained considerable traction and has begun to be tested.2,3,6,65 In this context, melphalan has been used in combination with adoptive T-cell therapy and cancer vaccines in the treatment of multiple myeloma. Rapoport et al. reported that in myeloma patients treated with HDM and ASCT, infusion of in vivo vaccine-primed, ex vivo costimulated autologous T cells followed by post-transplant vaccines improved the severe immunodeficiency associated with high-dose chemotherapy and led to enhanced memory T-cell responses.66–68 In an attempt to generate marrow-tropic and tumor-specific T-cell populations suitable for ACT, Noonan et al. established a protocol in which ex vivo–activated and –expanded marrow-infiltrating lymphocytes were used for adoptive immunotherapy of multiple myeloma.69 Their recent work has further demonstrated the feasibility and efficacy of marrow-infiltrating lymphocyte-based ACT following HDM and ASCT.70
In recent years, adoptive T-cell therapy using T cells engineered to express selected TCRs or chimeric antigen receptors (CARs) has emerged as a promising cancer treatment option.63,64 Fueled by the encouraging outcomes of ACT in melanoma and B-cell malignancies, these approaches have been applied to multiple myeloma. In two recent clinical trials, the HDM and ASCT regimen was used in combination with infusion of autologous T cells engineered to express NY-ESO-1-specific TCR or CD19-targeting chimeric antigen receptor (CAR).71,72 In both studies, the engineered T cells were safe and able to traffic to marrow and exhibited clinical activity against myeloma.
B. Potential of Melphalan as Preparative Chemotherapy for Adoptive Immunotherapy
In the aforementioned clinical studies, melphalan, as part of the standard care protocol, has been integrated with cancer immunotherapies. Its potential immune-potentiating effect has not been a factor considered in the research design, even though it might contribute to the therapeutic outcome. The immunostimulatory features of melphalan unraveled by our study support the notion that melphalan can be used as an immune-potentiating anticancer drug similar to CTX.59 We tested this in animal models in which tumor-specific CD4+ T cells were transferred to tumor-bearing mice following melphalan or CTX pre-conditioning. We found that when mice were pre-treated with melphalan at the dose that causes lymphodepletion, immunogenic cell death and cytokine/chemokine surge, the transferred tumor-specific CD4+ T cells underwent robust clonal expansion and effector differentiation, giving rise to polyfunctional CD4+ effector cells. Notably, the combination of melphalan and CD4+ T-cell ACT was more efficacious than either treatment alone in prolonging the survival of mice with advanced B-cell lymphomas or colorectal tumors. Notably, although the majority of current ACT protocols use CD8+ T cells or unfractionated total T cells, mounting evidence indicates that CD4+ T cells can be a good cellular source of ACT.73–77 In fact, the use of tumor-reactive CD4+ T cells for ACT has become a reality in the clinic and has demonstrated efficacy in patients.78,79 Therefore, our demonstration of the efficacy of melphalan and CD4+ T-cell ACT is clinically relevant. Even though our study focused on the use of CD4+ T cells for ACT, it is conceivable that CD8+ T-cell ACT will benefit from melphalan pre-conditioning as well. In summary, our study results imply that the immunomodulatory effects of melphalan can be exploited to potentiate the efficacy of adoptive T-cell therapy.
V. COUNTER-REGULATION MECHANISMS ASSOCIATED WITH MELPHALAN
A. Chemotherapy-Induced MDSCs Attenuate Antitumor Immune Responses
It has now been well-established that in addition to exerting direct cytotoxicity on fast growing tumor cells, many antineoplastic agents can incite antitumor immune responses. These anticancer drugs exert immunostimulatory effects through a variety of mechanisms.7,80 On the other hand, it also has been well-documented in animal models that chemotherapy can exert unwanted “opposite effects” that promote the growth of the residual tumors.81,82 This paradoxical feature of chemotherapy is exemplified by the alkylating agent CTX. With its well-recognized immunostimulatory effects, CTX has been widely used both in preclinical studies and clinical settings to enhance the efficacy of cancer immunotherapy such as adoptive T-cell therapy.9–11 Meanwhile, it has been shown that CTX is capable of promoting tumor metastasis,83,84 and inducing myeloid cells with immunosuppressive activities.85–90 We recently reported that CTX-induced myeloid cells can attenuate the efficacy of adoptive T-cell therapy in a PD1/PDL1-dependent fashion in a murine lymphoma model.91 Although the induction of inflammatory myeloid cells is not unique to CTX, a number of widely used anticancer drugs, including doxorubicin and paclitaxel, have been found to have similar effects.91–94 The induction of immunosuppressive MDSC-like myeloid cells by chemotherapy is not limited to animal models; chemotherapy-driven MDSC expansion has been observed in cancer patients. In a study conducted in patients with breast cancer, circulating MDSC numbers were significantly increased in patients receiving doxorubicin-CTX chemotherapy, and the results correlated with clinical cancer stage and metastatic tumor burden.95 Our recent study indicated that melphalan can also induce the expansion of inflammatory myeloid cells.91–94 Given that CTX-induced myeloid cells can be depleted or functionally blocked by low-dose gemcitabine, αCCR2 monoclonal antibody, or PD1/PDL1 blockade,91 the same approaches may be applicable to melphalan to inhibit its MDSC-promoting effect while preserving its immunostimulatory effects.
B. B7.1/CD28 Interaction Promotes the Survival of Myeloma
It has been shown that low-dose melphalan can induce B7.1 in tumor and stromal cells.56 Although this effect is considered immunogenic, it may also simultaneously contribute to tumor survival because myeloma cells express B7 receptor molecule CD28 on their surface. Upon ligation of CD28 on MM cells with B7 molecules on DCs (or by agonistic antibodies), tumor cells exhibited decreased apoptosis in response to chemotherapy or serum starvation.96 The interaction between MM and DCs triggers the production of IL-6 and indolamine-2,3-dioxygenase (IDO) by DCs.97 IL-6 can improve MM survival,98 whereas IDO promotes a tolerogenic milieu by suppressing effector T cells and activating regulatory T cells (Tregs).99 It is possible that melphalan induces B7.1 in tumor cells and stroma cells, which then engages CD28 on MM and hence promotes the survival and persistence of myeloma cells after chemotherapy. This notion is supported by the observation that CD28 expression on myeloma cells correlates with poor prognosis.100 Thus, it is conceivable that disrupting CD28-B7.1 interaction after melphalan treatment would be beneficial. Indeed, this was demonstrated by a recent study showing that blockade of CD28/B7 using CTLA4-Ig resulted in sensitization of myeloma cells to melphalan.101
C. IL-6 Signaling Pathway Promotes Tumor Drug Resistance
Multiple lines of evidence, from both animal and human studies, indicate that IL-6 is one of the cytokines markedly induced after melphalan treatment. 102,103 It is now greatly appreciated that enhanced IL-6/STAT3 signaling pathway is a common feature of human cancer leading to progression of the malignancy.104–106 IL-6 plays a role in hematopoiesis, immune regulation, and oncogenesis and influences cancer cell migration, invasion, metastatic potential, proliferation, angiogenesis, and apoptosis.107 IL-6 stimulates the growth of a number of cancers including MM, breast cancer, and lung cancer.108,109 Elevated levels of circulating IL-6 have been correlated with poor prognosis and multidrug resistance of tumor cells.110 IL-6 activates STAT3-dependent pathway and the signal mediated by IL-6/STAT3 is terminated by SOCS3.111 Upon translocation to the nucleus, pSTAT3 recognizes its DNA sequence called GAS (IFN-γ-activated sequence), which initiates changes in the expression of antiapoptotic molecules like Bcl2, Bcl-xL, c-Myc, or Fas. STAT3 also activates Ras, MEK, MAPK, Cox, and PI3K/Akt pathways.112 In MM, IL-6 can activate tumor cells in two ways: Akt independently (through RAS) and Akt dependently through PI3K p85/STAT3.113 IL-6 has also been attributed to the development of stem cell-like properties of tumor cells (cancer stem cells, CSC);114 IL-6 was shown to induce CSC sphere formation of MM cells by upregulation of Notch-dependent Jagged-1 in a STAT3-dependent manner.115 Therefore, disrupting the IL-6 signaling pathway represents an attractive strategy for cancer treatment.116 In line with this hypothesis, the combination of melphalan and siltuximab was able to overcome protective effect of bone marrow stroma on MM cells by inhibiting the PI3K/Akt pathway responsible for myeloma resistance to melphalan.117 Moreover, blockade of interleukin-6 signaling with siltuximab enhanced melphalan cytotoxicity in preclinical models of multiple myeloma.118 The combination of αIL-6 monoclonal antibody with MM standard care is currently being evaluated clinically.119–121
VI. CANCER CELLS ADAPT METABOLICALLY IN RESPONSE TO CHEMOTHERAPY
It has been well established that cancer cells can reprogram their metabolisms to meet the biosynthetic needs of rapid proliferation.122,123 There is emerging evidence that cancer cells can adapt metabolically in response to the stress elicited by chemical insults.124–126 Using transcriptomic and proteomic approaches, Zub et al. observed that in melphalan-resistant myeloma cell lines, there is a metabolic switch conforming to the Warburg effect (aerobic glycolysis) and an elevated oxidative stress response, resulting in enhanced survival and proliferation partially mediated by VEGF/IL-8 signaling.127 Specifically, in melphalan-resistant myeloma cell line RPMI8226, most of the glycolytic and pentose phosphate–pathway enzymes were upregulated, whereas the tricarboxylic acid cycle and electron transport chain proteins were downregulated. The switch of metabolic dependence to aerobic glycolysis implicates the glycolytic pathway as a potential target for therapeutic intervention to improve the efficacy of chemotherapy. Supporting this notion, inhibitors of selected metabolic and oxidative stress response enzymes displayed a selective cytotoxicity against melphalan-resistant myeloma cells. It was recently shown that metabolic competition, especially for glucose (and perhaps for other metabolites) in the tumor microenvironment, renders T cells dysfunctional even in the face of antigenic stimulation.128 Glucose consumption by tumor cells leads to glucose deprivation and inhibition of mTOR pathway in T cells, blocking T-cell effector differentiation.128 At the time of activation, CD28 signaling in T cells not only serves as a co-stimulation signal to fully activate T cells but also functions to increase T-cell glucose uptake and glycolysis during an immune response.129 On the other hand, immunosuppressive Tregs and M2 macrophages preferentially use fatty acids, rather than glucose, as an energy source. Therefore, in the tumor microenvironment, immunoregulatory cells are less likely to be affected by a tumor’s glucose consumption, leaving effector T cells most vulnerable to the reduced availability of glucose.130,131 Interestingly, Chang et al. showed that in immune checkpoint blockade targeting CTLA-4, PD1 and PDL1 can relieve the nutrient restriction on T effector cells and restore glucose availability in the tumor tissue,128 suggesting that the efficacy of immune checkpoint blockade is at least partially attributable to metabolic normalization of the tumor microenvironment.
VII. CONCLUSIONS
Melphalan has been used in the clinic for its potent antineoplastic activity. High-dose melphalan followed by autologous hematopoietic stem cell transplantation is regarded as a standard treatment for patients with multiple myeloma. Melphalan is also the mainstay of various chemotherapy regimens used for transplant-ineligible MM patients. In recent years, there have been multiple clinical trials in which melphalan was used in combination with adoptive T-cell therapy for refractory myeloma.65–67,69–71 This form of chemo-immunotherapy essentially applies immunotherapy on the platform of conventional chemotherapy in which melphalan is a major component. In these complex clinical studies, it is difficult to assess the contribution of the immunopotentiating effects of melphalan. Our animal study manifests the immunostimulatory effects of melphalan in a physiologically relevant setting.72 The ability of melphalan to augment CD4+ T-cell adoptive therapy provides a strong rationale for exploiting the immunopotentiating effects of melphalan in future clinical studies. Our study presents clear evidence that melphalan can induce immunogenic cell death, enhance tumor antigen uptake by DCs, eliminate Tregs, and induce transient lymphopenia along with a burst of proinflammatory cytokines/chemokines. Future studies should determine how each of these activities contribute to the enhanced efficacy of antitumor CD4+ T cells. Notably, the immunomodulatory effects of melphalan are not always beneficial or desirable. For example, melphalan-induced IL-6 and immunosuppressive myeloid cells can serve as counter-regulation mechanisms that attenuate the efficacy of immunotherapy. Thus, it will be important to take the necessary measures, such as IL-6 neutralization and PD1 blockade, to overcome the unfavorable tumor-promoting effects associated with melphalan. The results from our study support a scenario in which melphalan-induced HMGB1 attracts immature DCs into the draining lymph nodes; meanwhile, surface CRT expression on tumor cells presents an “eat-me” signal for phagocytosis by immature DCs, and the pro-inflammatory milieu induced by melphalan enhances DC maturation and antigen presentation. These events collectively contribute to robust CD4+ T-cell activation and effector differentiation. Once activated, polyfunctional CD4+ effector cells can mount immune responses against tumor cells through multiple mechanisms.132 In summary, our study identifies melphalan as an anticancer drug with potent immunopotentiating properties, and our results support its usefulness beyond its direct tumoricidal activity in the setting of chemo-immunotherapy for synergistic therapeutic outcomes.
Acknowledgments
The authors thank members of the Zhou Laboratory for insightful discussions. This work is funded by National Institutes of Health grant R01CA158202 and the American Cancer Society Research Scholar Grant (RSG-12-169-01-LIB) to G.Z.
ABBREVIATIONS
- ACT
adoptive T-cell therapy
- ASCT
autologous stem cell transplantation
- CRT
calreticulin
- CTX
cyclophosphamide
- DC
dendritic cells
- HDM
high dose melphalan
- ICD
immunogenic cell death
- LDM
low dose melphalan
- MDCSs
myeloid derived suppressor cells
- MM
multiple myeloma
- Tregs
regulatory T cells
References
- 1.Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of age. Nature. 2011;480(7378):480–9. doi: 10.1038/nature10673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Emens LA, Silverstein SC, Khleif S, Marincola FM, Galon J. Toward integrative cancer immunotherapy: targeting the tumor microenvironment. J Transl Med. 2012;10:70. doi: 10.1186/1479-5876-10-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol Res. 2015;3(5):436–43. doi: 10.1158/2326-6066.CIR-15-0064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Emens LA, Jaffee EM. Leveraging the activity of tumor vaccines with cytotoxic chemotherapy. Cancer Res. 2005;65(18):8059–64. doi: 10.1158/0008-5472.CAN-05-1797. [DOI] [PubMed] [Google Scholar]
- 5.Lake RA, Robinson BW. Immunotherapy and chemotherapy—a practical partnership. Nat Rev Cancer. 2005;5(5):397–405. doi: 10.1038/nrc1613. [DOI] [PubMed] [Google Scholar]
- 6.Prendergast GC, Jaffee EM. Cancer immunologists and cancer biologists: why we didn’t talk then but need to now. Cancer Res. 2007;67(8):3500–4. doi: 10.1158/0008-5472.CAN-06-4626. [DOI] [PubMed] [Google Scholar]
- 7.Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8(1):59–73. doi: 10.1038/nri2216. [DOI] [PubMed] [Google Scholar]
- 8.Haynes NM, van der Most RG, Lake RA, Smyth MJ. Immunogenic anti–cancer chemotherapy as an emerging concept. Curr Opin Immunol. 2008;20(5):545–57. doi: 10.1016/j.coi.2008.05.008. [DOI] [PubMed] [Google Scholar]
- 9.Klebanoff CA, Khong HT, Antony PA, Palmer DC, Restifo NP. Sinks, suppressors and antigen presenters: how lymphodepletion enhances T cell-mediated tumor immunotherapy. Trends Immunol. 2005;26(2):111–7. doi: 10.1016/j.it.2004.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol. 2011;33(4):369–83. doi: 10.1007/s00281-011-0245-0. [DOI] [PubMed] [Google Scholar]
- 11.Proietti E, Moschella F, Capone I, Belardelli F. Exploitation of the propulsive force of chemotherapy for improving the response to cancer immunotherapy. Molec Oncol. 2012;6(1):1–14. doi: 10.1016/j.molonc.2011.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gilman A, Philips FS. The biological actions and therapeutic applications of the b-chloroethyl amines and sulfides. Science. 1946;103(2675):409–15. [PubMed] [Google Scholar]
- 13.Bayraktar UD, Bashir Q, Qazilbash M, Champlin RE, Ciurea SO. Fifty years of melphalan use in hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19(3):344–56. doi: 10.1016/j.bbmt.2012.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Begleiter A, Lam HY, Grover J, Froese E, Goldenberg GJ. Evidence for active transport of melphalan by two amino acid carriers in l5178y lymphoblasts in vitro. Cancer Res. 1979;39(2 Pt 1):353–9. [PubMed] [Google Scholar]
- 15.Samuels BL, Bitran JD. High-dose intravenous melphalan: a review. J Clin Oncol. 1995;13(7):1786–99. doi: 10.1200/JCO.1995.13.7.1786. [DOI] [PubMed] [Google Scholar]
- 16.Povirk LF, Shuker DE. DNA damage and mutagenesis induced by nitrogen mustards. Mutat Res. 1994;318(3):205–26. doi: 10.1016/0165-1110(94)90015-9. [DOI] [PubMed] [Google Scholar]
- 17.Ahmed AE, Hsu TF, el-Azhary RA, Moawad H, Farrish HH, Jr, Costanzi J. Tissue distribution and macromolecular interactions of 14[c-ring] melphalan in the rat. Cancer Chemother Pharmacol. 1982;8(3):271–6. doi: 10.1007/BF00254049. [DOI] [PubMed] [Google Scholar]
- 18.Sarosy G, Leyland-Jones B, Soochan P, Cheson BD. The systemic administration of intravenous melphalan. J Clin Oncol. 1988;6(11):1768–82. doi: 10.1200/JCO.1988.6.11.1768. [DOI] [PubMed] [Google Scholar]
- 19.Smith ML, Newland AC. Treatment of myeloma. QJM. 1999;92(1):11–4. doi: 10.1093/qjmed/92.1.11. [DOI] [PubMed] [Google Scholar]
- 20.Gregory WM, Richards MA, Malpas JS. Combination chemotherapy versus melphalan and prednisolone in the treatment of multiple myeloma: an overview of published trials. J Clin Oncol. 1992;10(2):334–42. doi: 10.1200/JCO.1992.10.2.334. [DOI] [PubMed] [Google Scholar]
- 21.Dredge K, Marriott JB, Macdonald CD, Man HW, Chen R, Muller GW, Stirling D, Dalgleish AG. Novel thalidomide analogues display anti-angiogenic activity independently of immunomodulatory effects. Brit J Cancer. 2002;87(10):1166–72. doi: 10.1038/sj.bjc.6600607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Singhal S, Mehta J, Desikan R, Ayers D, Roberson P, Eddlemon P, Munshi N, Anaissie E, Wilson C, Dhodapkar M, Zeddis J, Barlogie B. Antitumor activity of thalidomide in refractory multiple myeloma. New Engl J Med. 1999;341(21):1565–71. doi: 10.1056/NEJM199911183412102. [DOI] [PubMed] [Google Scholar]
- 23.Kapoor P, Ramakrishnan V, Rajkumar SV. Bortezomib combination therapy in multiple myeloma. Semin Hematol. 2012;49(3):228–42. doi: 10.1053/j.seminhematol.2012.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Stewart AK. Medicine. How thalidomide works against cancer. Science. 2014;343(6168):256–7. doi: 10.1126/science.1249543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS, Dmoszynska A, Abdulkadyrov KM, Schots R, Jiang B, Mateos MV, Anderson KC, Esseltine DL, Liu K, Cakana A, van de Velde H, Richardson PG Investigators VT. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. New Engl J Med. 2008;359(9):906–17. doi: 10.1056/NEJMoa0801479. [DOI] [PubMed] [Google Scholar]
- 26.Palumbo A, Ambrosini MT, Benevolo G, Pregno P, Pescosta N, Callea V, Cangialosi C, Caravita T, Morabito F, Musto P, Bringhen S, Falco P, Avonto I, Cavallo F, Boccadoro M Italian Multiple Myeloma N, Gruppo Italiano Malattie Ematologicche dA. Bortezomib, melphalan, prednisone, and thalidomide for relapsed multiple myeloma. Blood. 2007;109(7):2767–72. doi: 10.1182/blood-2006-08-042275. [DOI] [PubMed] [Google Scholar]
- 27.Mohty M, Harousseau JL. Treatment of autologous stem cell transplant-eligible multiple myeloma patients: ten questions and answers. Haematologica. 2014;99(3):408–16. doi: 10.3324/haematol.2013.096149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Palumbo A, Rajkumar SV. Treatment of newly diagnosed myeloma. Leukemia. 2009;23(3):449–56. doi: 10.1038/leu.2008.325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jantunen E, Kuittinen T, Penttila K, Lehtonen P, Mahlamaki E, Nousiainen T. High-dose melphalan (200 mg/m2) supported by autologous stem cell transplantation is safe and effective in elderly (>or=65 years) myeloma patients: comparison with younger patients treated on the same protocol. Bone Marrow Transplant. 2006;37(10):917–22. doi: 10.1038/sj.bmt.1705360. [DOI] [PubMed] [Google Scholar]
- 30.Mutlu P, Ural AU, Gunduz U. Different types of cell cycle- and apoptosis-related gene expressions alter in corticosteroid-, vincristine-, and melphalan-resistant U-266 multiple myeloma cell lines. Turk J Hematol. 2014;31(3):231–8. doi: 10.4274/tjh.2013.0231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Park GB, Kim YS, Kim D, Kim S, Lee HK, Cho DH, Lee WJ, Hur DY. Melphalan-induced apoptosis of EBV-transformed B cells through upregulation of TAP73 and XAF1 and nuclear import of XPA. J Immunol. 2013;191(12):6281–91. doi: 10.4049/jimmunol.1203442. [DOI] [PubMed] [Google Scholar]
- 32.Shaw PJ, Nath CE, Lazarus HM. Not too little, not too much-just right! (better ways to give high dose melphalan) Bone Marrow Transplant. 2014;49(12):1457–65. doi: 10.1038/bmt.2014.186. [DOI] [PubMed] [Google Scholar]
- 33.Gorin NC, Giebel S, Labopin M, Savani BN, Mohty M, Nagler A. Autologous stem cell transplantation for adult acute leukemia in 2015: Time to rethink? Present status and future prospects. Bone Marrow Transplant. 2015;50(12):1495–502. doi: 10.1038/bmt.2015.179. [DOI] [PubMed] [Google Scholar]
- 34.Herbers AH, de Haan AF, van der Velden WJ, Donnelly JP, Blijlevens NM. Mucositis not neutropenia determines bacteremia among hematopoietic stem cell transplant recipients. Transplant Infect Dis. 2014;16(2):279–85. doi: 10.1111/tid.12195. [DOI] [PubMed] [Google Scholar]
- 35.Nooka AK, Harvey RD, Langston A, Collins H, Lonial S, Kaufman JL. Optimal dosing of melphalan as high-dose therapy before autologous hematopoietic stem cell transplantation in myeloma patients with solitary kidney: a case series. Clin Lymphoma Myeloma Leukemia. 2014;14(2):e59–63. doi: 10.1016/j.clml.2013.11.007. [DOI] [PubMed] [Google Scholar]
- 36.Dunkel IJ, Shi W, Salvaggio K, Marr BP, Brodie SE, Gobin YP, Abramson DH. Risk factors for severe neutropenia following intra-arterial chemotherapy for intra-ocular retinoblastoma. PloS One. 2014;9(10):e108692. doi: 10.1371/journal.pone.0108692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Condomines M, Veyrune JL, Larroque M, Quittet P, Latry P, Lugagne C, Hertogh C, Kanouni T, Rossi JF, Klein B. Increased plasma-immune cytokines throughout the high-dose melphalan-induced lymphodepletion in patients with multiple myeloma: a window for adoptive immunotherapy. J Immunol. 2010;184(2):1079–84. doi: 10.4049/jimmunol.0804159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5(4):263–74. doi: 10.1038/nrc1586. [DOI] [PubMed] [Google Scholar]
- 39.Dudek-Peric AM, Golab J, Garg AD, Agostinis P. Melanoma targeting with the loco-regional chemotherapeutic, melphalan: from cell death to immunotherapeutic efficacy. Oncoimmunology. 2015;4(12):e1054600. doi: 10.1080/2162402X.2015.1054600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, Coutant F, Metivier D, Pichard E, Aucouturier P, Pierron G, Garrido C, Zitvogel L, Kroemer G. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exper Med. 2005;202(12):1691–701. doi: 10.1084/jem.20050915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A, Mignot G, Maiuri MC, Ullrich E, Saulnier P, Yang H, Amigorena S, Ryffel B, Barrat FJ, Saftig P, Levi F, Lidereau R, Nogues C, Mira JP, Chompret A, Joulin V, Clavel-Chapelon F, Bourhis J, Andre F, Delaloge S, Tursz T, Kroemer G, Zitvogel L. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13(9):1050–9. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
- 42.Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, Metivier D, Larochette N, van Endert P, Ciccosanti F, Piacentini M, Zitvogel L, Kroemer G. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13(1):54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
- 43.Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E, Perfettini JL, Schlemmer F, Tasdemir E, Uhl M, Genin P, Civas A, Ryffel B, Kanellopoulos J, Tschopp J, Andre F, Lidereau R, McLaughlin NM, Haynes NM, Smyth MJ, Kroemer G, Zitvogel L. Activation of the nlrp3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat Med. 2009;15(10):1170–8. doi: 10.1038/nm.2028. [DOI] [PubMed] [Google Scholar]
- 44.Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, Sanchez M, Lorenzi S, D’Urso MT, Belardelli F, Gabriele L, Proietti E, Bracci L. Cyclophosphamide synergizes with type iinterferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res. 2011;71(3):768–78. doi: 10.1158/0008-5472.CAN-10-2788. [DOI] [PubMed] [Google Scholar]
- 45.Lutsiak ME, Semnani RT, De Pascalis R, Kashmiri SV, Schlom J, Sabzevari H. Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide. Blood. 2005;105(7):2862–8. doi: 10.1182/blood-2004-06-2410. [DOI] [PubMed] [Google Scholar]
- 46.van der Most RG, Currie AJ, Mahendran S, Prosser A, Darabi A, Robinson BW, Nowak AK, Lake RA. Tumor eradication after cyclophosphamide depends on concurrent depletion of regulatory t cells: a role for cycling TNFR2-expressing effector-suppressor t cells in limiting effective chemotherapy. Cancer Immunol Immunother. 2009;58(8):1219–28. doi: 10.1007/s00262-008-0628-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kuczma M, Kopij M, Pawlikowska I, Wang CY, Rempala GA, Kraj P. Intratumoral convergence of the tcr repertoires of effector and Foxp3+ CD4+ T cells. PloS One. 2010;5(10):e13623. doi: 10.1371/journal.pone.0013623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Barbi J, Pardoll D, Pan F. Treg functional stability and its responsiveness to the microenvironment. Immunol Rev. 2014;259(1):115–39. doi: 10.1111/imr.12172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11(18):6713–21. doi: 10.1158/1078-0432.CCR-05-0883. [DOI] [PubMed] [Google Scholar]
- 50.Ozao-Choy J, Ma G, Kao J, Wang GX, Meseck M, Sung M, Schwartz M, Divino CM, Pan PY, Chen SH. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res. 2009;69(6):2514–22. doi: 10.1158/0008-5472.CAN-08-4709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Alizadeh D, Trad M, Hanke NT, Larmonier CB, Janikashvili N, Bonnotte B, Katsanis E, Larmonier N. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74(1):104–18. doi: 10.1158/0008-5472.CAN-13-1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, Martin F, Apetoh L, Rebe C, Ghiringhelli F. 5-fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70(8):3052–61. doi: 10.1158/0008-5472.CAN-09-3690. [DOI] [PubMed] [Google Scholar]
- 53.Kodumudi KN, Woan K, Gilvary DL, Sahakian E, Wei S, Djeu JY. A novel chemoimmunomodulating property of docetaxel: suppression of myeloid-derived suppressor cells in tumor bearers. Clin Cancer Res. 2010;16(18):4583–94. doi: 10.1158/1078-0432.CCR-10-0733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Moschella F, Valentini M, Arico E, Macchia I, Sestili P, D’Urso MT, Alessandri C, Belardelli F, Proietti E. Unraveling cancer chemoimmunotherapy mechanisms by gene and protein expression profiling of responses to cyclophosphamide. Cancer Res. 2011;71(10):3528–39. doi: 10.1158/0008-5472.CAN-10-4523. [DOI] [PubMed] [Google Scholar]
- 55.Bocian RC, Ben-Efraim S, Dray S, Mokyr MB. Melphalan-mediated potentiation of antitumor immune responsiveness of immunosuppressed spleen cells from mice bearing a large MOPC-315 tumor. Cancer Immunol Immunother. 1984;18(1):41–8. doi: 10.1007/BF00205398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sojka DK, Donepudi M, Bluestone JA, Mokyr MB. Melphalan and other anticancer modalities up-regulate B7-1 gene expression in tumor cells. J Immunol. 2000;164(12):6230–6. doi: 10.4049/jimmunol.164.12.6230. [DOI] [PubMed] [Google Scholar]
- 57.Tongu M, Harashima N, Monma H, Inao T, Yamada T, Kawauchi H, Harada M. Metronomic chemotherapy with low-dose cyclophosphamide plus gemcitabine can induce anti-tumor T cell immunity in vivo. Cancer Immunol Immunother. 2013;62(2):383–91. doi: 10.1007/s00262-012-1343-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Bracci L, Moschella F, Sestili P, La Sorsa V, Valentini M, Canini I, Baccarini S, Maccari S, Ramoni C, Belardelli F, Proietti E. Cyclophosphamide enhances the antitumor efficacy of adoptively transferred immune cells through the induction of cytokine expression, B-cell and T-cell homeostatic proliferation, and specific tumor infiltration. Clin Cancer Res. 2007;13(2 Pt 1):644–53. doi: 10.1158/1078-0432.CCR-06-1209. [DOI] [PubMed] [Google Scholar]
- 59.Lu X, Ding ZC, Cao Y, Liu C, Habtetsion T, Yu M, Lemos H, Salman H, Xu H, Mellor AL, Zhou G. Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor-specific CD4+ T cells. J Immunol. 2015;194(4):2011–21. doi: 10.4049/jimmunol.1401894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dudek-Peric AM, Ferreira GB, Muchowicz A, Wouters J, Prada N, Martin S, Kiviluoto S, Winiarska M, Boon L, Mathieu C, van den Oord J, Stas M, Gougeon ML, Golab J, Garg AD, Agostinis P. Antitumor immunity triggered by melphalan is potentiated by melanoma cell surface-associated calreticulin. Cancer Res. 2015;75(8):1603–14. doi: 10.1158/0008-5472.CAN-14-2089. [DOI] [PubMed] [Google Scholar]
- 61.Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Snyder N, Sarkar S. Drug resistance in cancer: an overview. Cancers (Basel) 2014;6(3):1769–92. doi: 10.3390/cancers6031769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161(2):205–14. doi: 10.1016/j.cell.2015.03.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013;39(1):49–60. doi: 10.1016/j.immuni.2013.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62–8. doi: 10.1126/science.aaa4967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Emens LA, Machiels JP, Reilly RT, Jaffee EM. Chemotherapy: friend or foe to cancer vaccines? Curr Opin Molec Therapeut. 2001;3(1):77–84. [PubMed] [Google Scholar]
- 66.Rapoport AP, Stadtmauer EA, Aqui N, Badros A, Cotte J, Chrisley L, Veloso E, Zheng Z, Westphal S, Mair R, Chi N, Ratterree B, Pochran MF, Natt S, Hinkle J, Sickles C, Sohal A, Ruehle K, Lynch C, Zhang L, Porter DL, Luger S, Guo C, Fang HB, Blackwelder W, Hankey K, Mann D, Edelman R, Frasch C, Levine BL, Cross A, June CH. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med. 2005;11(11):1230–7. doi: 10.1038/nm1310. [DOI] [PubMed] [Google Scholar]
- 67.Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Xu YY, Kalos M, Cai L, Fang HB, Weiss BM, Badros A, Yanovich S, Akpek G, Tsao P, Cross A, Mann D, Philip S, Kerr N, Brennan A, Zheng Z, Ruehle K, Milliron T, Strome SE, Salazar AM, Levine BL, June CH. Combination immunotherapy after asct for multiple myeloma using MAGE-A3/Poly-ICLC immunizations followed by adoptive transfer of vaccine-primed and costimulated autologous t cells. Clin Cancer Res. 2014;20(5):1355–65. doi: 10.1158/1078-0432.CCR-13-2817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, Janofsky S, Chew A, Storek J, Akpek G, Badros A, Yanovich S, Tan MT, Veloso E, Pasetti MF, Cross A, Philip S, Murphy H, Bhagat R, Zheng Z, Milliron T, Cotte J, Cannon A, Levine BL, Vonderheide RH, June CH. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of htert and survivin after ASCT for myeloma. Blood. 2011;117(3):788–97. doi: 10.1182/blood-2010-08-299396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Noonan K, Matsui W, Serafini P, Carbley R, Tan G, Khalili J, Bonyhadi M, Levitsky H, Whartenby K, Borrello I. Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer Res. 2005;65(5):2026–34. doi: 10.1158/0008-5472.CAN-04-3337. [DOI] [PubMed] [Google Scholar]
- 70.Noonan KA, Huff CA, Davis J, Lemas MV, Fiorino S, Bitzan J, Ferguson A, Emerling A, Luznik L, Matsui W, Powell J, Fuchs E, Rosner GL, Epstein C, Rudraraju L, Ambinder RF, Jones RJ, Pardoll D, Borrello I. Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci Translat Med. 2015;7(288):288ra78. doi: 10.1126/scitranslmed.aaa7014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, Badros AZ, Garfall A, Weiss B, Finklestein J, Kulikovskaya I, Sinha SK, Kronsberg S, Gupta M, Bond S, Melchiori L, Brewer JE, Bennett AD, Gerry AB, Pumphrey NJ, Williams D, Tayton-Martin HK, Ribeiro L, Holdich T, Yanovich S, Hardy N, Yared J, Kerr N, Philip S, Westphal S, Siegel DL, Levine BL, Jakobsen BK, Kalos M, June CH. NYESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21(8):914–21. doi: 10.1038/nm.3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ, Zheng Z, Vogl DT, Cohen AD, Weiss BM, Dengel K, Kerr ND, Bagg A, Levine BL, June CH, Stadtmauer EA. Chimeric antigen receptor T cells against CD19 for multiple myeloma. New Engl J Med. 2015;373(11):1040–7. doi: 10.1056/NEJMoa1504542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Greenberg PD, Kern DE, Cheever MA. Therapy of disseminated murine leukemia with cyclophosphamide and immune Lyt-1+,2- T cells. Tumor eradication does not require participation of cytotoxic T cells. Journal of Experimental Medicine. 1985;161(5):1122–34. doi: 10.1084/jem.161.5.1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Corthay A, Skovseth DK, Lundin KU, Rosjo E, Omholt H, Hofgaard PO, Haraldsen G, Bogen B. Primary antitumor immune response mediated by CD4+ T cells. Immunity. 2005;22(3):371–83. doi: 10.1016/j.immuni.2005.02.003. [DOI] [PubMed] [Google Scholar]
- 75.Perez-Diez A, Joncker NT, Choi K, Chan WF, Anderson CC, Lantz O, Matzinger P. CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood. 2007;109(12):5346–54. doi: 10.1182/blood-2006-10-051318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Quezada SA, Simpson TR, Peggs KS, Merghoub T, Vider J, Fan X, Blasberg R, Yagita H, Muranski P, Antony PA, Restifo NP, Allison JP. Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J Exper Med. 2010;207(3):637–50. doi: 10.1084/jem.20091918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Muranski P, Restifo NP. Adoptive immunotherapy of cancer using CD4(+) T cells. Curr Opin Immunol. 2009;21(2):200–8. doi: 10.1016/j.coi.2009.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, Jungbluth A, Gnjatic S, Thompson JA, Yee C. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. New Engl J Med. 2008;358(25):2698–703. doi: 10.1056/NEJMoa0800251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP, Hogan K, Hinrichs CS, Parkhurst MR, Yang JC, Rosenberg SA. Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science. 2014;344(6184):641–5. doi: 10.1126/science.1251102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zitvogel L, Kepp O, Kroemer G. Immune parameters affecting the efficacy of chemotherapeutic regimens. Nat Rev Clin Oncol. 2011;8(3):151–60. doi: 10.1038/nrclinonc.2010.223. [DOI] [PubMed] [Google Scholar]
- 81.Daenen LG, Houthuijzen JM, Cirkel GA, Roodhart JM, Shaked Y, Voest EE. Treatment- induced host-mediated mechanisms reducing the efficacy of antitumor therapies. Oncogene. 2014;33(11):1341–7. doi: 10.1038/onc.2013.94. [DOI] [PubMed] [Google Scholar]
- 82.Ebos JM. Prodding the beast: assessing the impact of treatment-induced metastasis. Cancer Res. 2015;75(17):3427–35. doi: 10.1158/0008-5472.CAN-15-0308. [DOI] [PubMed] [Google Scholar]
- 83.Carmel RJ, Brown JM. The effect of cyclophosphamide and other drugs on the incidence of pulmonary metastases in mice. Cancer Res. 1977;37(1):145–51. [PubMed] [Google Scholar]
- 84.Yamauchi K, Yang M, Hayashi K, Jiang P, Yamamoto N, Tsuchiya H, Tomita K, Moossa AR, Bouvet M, Hoffman RM. Induction of cancer metastasis by cyclophosphamide pretreatment of host mice: an opposite effect of chemotherapy. Cancer Res. 2008;68(2):516–20. doi: 10.1158/0008-5472.CAN-07-3063. [DOI] [PubMed] [Google Scholar]
- 85.McIntosh KR, Segre M, Segre D. Characterization of cyclophosphamide-induced suppressor cells. Immunopharmacology. 1982;4(4):279–89. doi: 10.1016/0162-3109(82)90049-2. [DOI] [PubMed] [Google Scholar]
- 86.Segre M, Tomei E, Segre D. Cyclophosphamide-induced suppressor cells in mice: suppression of the antibody response in vitro and characterization of the effector cells. Cell Immunol. 1985;91(2):443–54. doi: 10.1016/0008-8749(85)90242-4. [DOI] [PubMed] [Google Scholar]
- 87.Nikcevich DA, Duffie GP, Young MR, Ellis NK, Kaufman GE, Wepsic HT. Stimulation of suppressor cells in the bone marrow and spleens of high dose cyclophosphamide-treated C57BL/6 mice. Cell Immunol. 1987;109(2):349– 59. doi: 10.1016/0008-8749(87)90318-2. [DOI] [PubMed] [Google Scholar]
- 88.Brooks-Kaiser JC, Bourque LA, Hoskin DW. Heterogeneity of splenic natural suppressor cells induced in mice by treatment with cyclophosphamide. Immunopharmacology. 1993;25(2):117–29. doi: 10.1016/0162-3109(93)90015-i. [DOI] [PubMed] [Google Scholar]
- 89.Angulo I, de las Heras FG, Garcia-Bustos JF, Gargallo D, Munoz-Fernandez MA, Fresno M. Nitric oxide-producing CD11b(+)Ly-6g(Gr-1)(+)CD31(ER-MP12)(+) cells in the spleen of cyclophosphamide-treated mice: implications for T-cell responses in immunosuppressed mice. Blood. 2000;95(1):212–20. [PubMed] [Google Scholar]
- 90.Mikyskova R, Indrova M, Pollakova V, Bieblova J, Simova J, Reinis M. Cyclophosphamide-induced myeloid-derived suppressor cell population is immunosuppressive but not identical to myeloid-derived suppressor cells induced by growing TC-1 tumors. J Immunother. 2012;35(5):374–84. doi: 10.1097/CJI.0b013e318255585a. [DOI] [PubMed] [Google Scholar]
- 91.Ding ZC, Lu X, Yu M, Lemos H, Huang L, Chandler P, Liu K, Walters M, Krasinski A, Mack M, Blazar BR, Mellor AL, Munn DH, Zhou G. Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T-cell responses through the PD-1-PD-L1 axis. Cancer Res. 2014;74(13):3441–53. doi: 10.1158/0008-5472.CAN-13-3596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lu X, Ding Z, Cao Y, Liu C, Habtetsion T, Yu M, Lemos H, Salman H, Xu H, Mellor AL, Zhou G. Alkylating agent melphalan augments the efficacy of adoptive immunotherapy using tumor-specific CD4+ T cells. J Immunol. 2015 doi: 10.4049/jimmunol.1401894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Nakasone ES, Askautrud HA, Kees T, Park JH, Plaks V, Ewald AJ, Fein M, Rasch MG, Tan YX, Qiu J, Park J, Sinha P, Bissell MJ, Frengen E, Werb Z, Egeblad M. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell. 2012;21(4):488–503. doi: 10.1016/j.ccr.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Volk-Draper L, Hall K, Griggs C, Rajput S, Kohio P, DeNardo D, Ran S. Paclitaxel therapy promotes breast cancer metastasis in a TLR4-dependent manner. Cancer Res. 2014;74(19):5421–34. doi: 10.1158/0008-5472.CAN-14-0067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Diaz-Montero CM, Salem ML, Nishimura MI, Garrett-Mayer E, Cole DJ, Montero AJ. Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2009;58(1):49–59. doi: 10.1007/s00262-008-0523-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Bahlis NJ, King AM, Kolonias D, Carlson LM, Liu HY, Hussein MA, Terebelo HR, Byrne GE, Jr, Levine BL, Boise LH, Lee KP. CD28-mediated regulation of multiple myeloma cell proliferation and survival. Blood. 2007;109(11):5002–10. doi: 10.1182/blood-2006-03-012542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Nair JR, Carlson LM, Koorella C, Rozanski CH, Byrne GE, Bergsagel PL, Shaughnessy JP, Jr, Boise LH, Chanan-Khan A, Lee KP. CD28 expressed on malignant plasma cells induces a prosurvival and immunosuppressive microenvironment. J Immunol. 2011;187(3):1243–53. doi: 10.4049/jimmunol.1100016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hulkkonen J, Pertovaara M, Antonen J, Pasternack A, Hurme M. Elevated interleukin-6 plasma levels are regulated by the promoter region polymorphism of the IL6 gene in primary Sjogren’s syndrome and correlate with the clinical manifestations of the disease. Rheumatology (Oxford) 2001;40(6):656–61. doi: 10.1093/rheumatology/40.6.656. [DOI] [PubMed] [Google Scholar]
- 99.Sharma MD, Hou DY, Liu Y, Koni PA, Metz R, Chandler P, Mellor AL, He Y, Munn DH. Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to Th17-like cells in tumor-draining lymph nodes. Blood. 2009;113(24):6102–11. doi: 10.1182/blood-2008-12-195354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Robillard N, Jego G, Pellat-Deceunynck C, Pineau D, Puthier D, Mellerin MP, Barille S, Rapp MJ, Harousseau JL, Amiot M, Bataille R. CD28, a marker associated with tumoral expansion in multiple myeloma. Clin Cancer Res. 1998;4(6):1521–6. [PubMed] [Google Scholar]
- 101.Murray ME, Gavile CM, Nair JR, Koorella C, Carlson LM, Buac D, Utley A, Chesi M, Bergsagel PL, Boise LH, Lee KP. CD28-mediated pro-survival signaling induces chemotherapeutic resistance in multiple myeloma. Blood. 2014;123(24):3770–9. doi: 10.1182/blood-2013-10-530964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chauhan D, Uchiyama H, Akbarali Y, Urashima M, Yamamoto K, Libermann TA, Anderson KC. Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa b. Blood. 1996;87(3):1104–12. [PubMed] [Google Scholar]
- 103.Lauta VM. A review of the cytokine network in multiple myeloma: diagnostic, prognostic, and therapeutic implications. Cancer. 2003;97(10):2440–52. doi: 10.1002/cncr.11072. [DOI] [PubMed] [Google Scholar]
- 104.Anglesio MS, George J, Kulbe H, Friedlander M, Rischin D, Lemech C, Power J, Coward J, Cowin PA, House CM, Chakravarty P, Gorringe KL, Campbell IG, Okamoto A, Birrer MJ, Huntsman DG, de Fazio A, Kalloger SE, Balkwill F, Gilks CB, Bowtell DD Australian Ovarian Cancer Study G. Il6-Stat3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Clin Cancer Res. 2011;17(8):2538–48. doi: 10.1158/1078-0432.CCR-10-3314. [DOI] [PubMed] [Google Scholar]
- 105.Marotta LL, Almendro V, Marusyk A, Shipitsin M, Schemme J, Walker SR, Bloushtain-Qimron N, Kim JJ, Choudhury SA, Maruyama R, Wu Z, Gonen M, Mulvey LA, Bessarabova MO, Huh SJ, Silver SJ, Kim SY, Park SY, Lee HE, Anderson KS, Richardson AL, Nikolskaya T, Nikolsky Y, Liu XS, Root DE, Hahn WC, Frank DA, Polyak K. The Jak2/Stat3 signaling pathway is required for growth of CD44(+)CD24(−) stem cell-like breast cancer cells in human tumors. J Clin Investig. 2011;121(7):2723–35. doi: 10.1172/JCI44745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wan S, Zhao E, Kryczek I, Vatan L, Sadovskaya A, Ludema G, Simeone DM, Zou W, Welling TH. Tumor-associated macrophages produce interleukin 6 and signal via Stat3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterol. 2014;147(6):1393–404. doi: 10.1053/j.gastro.2014.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC. Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer. 2007;7(8):585–98. doi: 10.1038/nrc2189. [DOI] [PubMed] [Google Scholar]
- 108.Jernberg H, Pettersson M, Kishimoto T, Nilsson K. Heterogeneity in response to interleukin 6 (Il-6), expression of Il-6 and Il-6 receptor mRNA in a panel of established human multiple myeloma cell lines. Leukemia. 1991;5(3):255–65. [PubMed] [Google Scholar]
- 109.Schafer ZT, Brugge JS. Il-6 involvement in epithelial cancers. J Clin Investig. 2007;117(12):3660–3. doi: 10.1172/JCI34237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wierzbowska A, Urbanska-Rys H, Robak T. Circulating IL-6-type cytokines and sIL-6R in patients with multiple myeloma. Brit J Haematol. 1999;105(2):412–9. [PubMed] [Google Scholar]
- 111.Croker BA, Krebs DL, Zhang JG, Wormald S, Willson TA, Stanley EG, Robb L, Greenhalgh CJ, Forster I, Clausen BE, Nicola NA, Metcalf D, Hilton DJ, Roberts AW, Alexander WS. SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol. 2003;4(6):540–5. doi: 10.1038/ni931. [DOI] [PubMed] [Google Scholar]
- 112.Carpenter RL, Lo HW. Stat3 target genes relevant to human cancers. Cancers (Basel) 2014;6(2):897–925. doi: 10.3390/cancers6020897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hideshima T, Nakamura N, Chauhan D, Anderson KC. Biologic sequelae of interleukin-6 induced PI3-K/AKT signaling in multiple myeloma. Oncogene. 2001;20(42):5991–6000. doi: 10.1038/sj.onc.1204833. [DOI] [PubMed] [Google Scholar]
- 114.Kim SY, Kang JW, Song X, Kim BK, Yoo YD, Kwon YT, Lee YJ. Role of the IL-6-JAK1-STAT3-OCT-4 pathway in the conversion of non-stem cancer cells into cancer stem-like cells. Cell Signal. 2013;25(4):961–9. doi: 10.1016/j.cellsig.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Yang Z, Guo L, Liu D, Sun L, Chen H, Deng Q, Liu Y, Yu M, Ma Y, Guo N, Shi M. Acquisition of resistance to trastuzumab in gastric cancer cells is associated with activation of Il-6/Stat3/Jagged-1/Notch positive feedback loop. Oncotarget. 2015;6(7):5072–87. doi: 10.18632/oncotarget.3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Trikha M, Corringham R, Klein B, Rossi JF. Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: a review of the rationale and clinical evidence. Clin Cancer Res. 2003;9(13):4653–65. [PMC free article] [PubMed] [Google Scholar]
- 117.Hideshima T, Catley L, Yasui H, Ishitsuka K, Raje N, Mitsiades C, Podar K, Munshi NC, Chauhan D, Richardson PG, Anderson KC. Perifosine, an oral bioactive novel alkylphospholipid, inhibits AKT and induces in vitro and in vivo cytotoxicity in human multiple myeloma cells. Blood. 2006;107(10):4053–62. doi: 10.1182/blood-2005-08-3434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hunsucker SA, Magarotto V, Kuhn DJ, Kornblau SM, Wang M, Weber DM, Thomas SK, Shah JJ, Voorhees PM, Xie H, Cornfeld M, Nemeth JA, Orlowski RZ. Blockade of interleukin-6 signalling with siltuximab enhances melphalan cytotoxicity in preclinical models of multiple myeloma. Brit J Haematol. 2011;152(5):579–92. doi: 10.1111/j.1365-2141.2010.08533.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rossi JF, Fegueux N, Lu ZY, Legouffe E, Exbrayat C, Bozonnat MC, Navarro R, Lopez E, Quittet P, Daures JP, Rouille V, Kanouni T, Widjenes J, Klein B. Optimizing the use of anti-interleukin-6 monoclonal antibody with dexamethasone and 140 mg/m2 of melphalan in multiple myeloma: results of a pilot study including biological aspects. Bone Marrow Transplant. 2005;36(9):771–9. doi: 10.1038/sj.bmt.1705138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Orlowski RZ, Gercheva L, Williams C, Sutherland H, Robak T, Masszi T, Goranova-Marinova V, Dimopoulos MA, Cavenagh JD, Spicka I, Maiolino A, Suvorov A, Blade J, Samoylova O, Puchalski TA, Reddy M, Bandekar R, van de Velde H, Xie H, Rossi JF. A phase 2, randomized, double-blind, placebo-controlled study of siltuximab (anti-Il-6 mAb) and bortezomib versus bortezomib alone in patients with relapsed or refractory multiple myeloma. Am J Hematol. 2015;90(1):42–9. doi: 10.1002/ajh.23868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Rossi JF, Negrier S, James ND, Kocak I, Hawkins R, Davis H, Prabhakar U, Qin X, Mulders P, Berns B. A phase i/ii study of siltuximab (cnto 328), an anti-interleukin-6 monoclonal antibody, in metastatic renal cell cancer. Brit J Cancer. 2010;103(8):1154–62. doi: 10.1038/sj.bjc.6605872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 123.Cairns RA, Harris IS, Mak TW. Regulation of cancer cell metabolism. Nat Rev Cancer. 2011;11(2):85–95. doi: 10.1038/nrc2981. [DOI] [PubMed] [Google Scholar]
- 124.Hess JA, Khasawneh MK. Cancer metabolism and oxidative stress: insights into carcinogenesis and chemotherapy via the non-dihydrofolate reductase effects of methotrexate. Biochim Biophys Acta Clin. 2015;3:152–61. doi: 10.1016/j.bbacli.2015.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Peiris-Pages M, Sotgia F, Lisanti MP. Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine hedgehog-gli signalling in breast cancer cells. Oncotarget. 2015;6(13):10728–45. doi: 10.18632/oncotarget.3828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Phan LM, Yeung SC, Lee MH. Cancer metabolic reprogramming: importance, main features, and potentials for precise targeted anti-cancer therapies. Cancer Biol Med. 2014;11(1):1–19. doi: 10.7497/j.issn.2095-3941.2014.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zub KA, Sousa MM, Sarno A, Sharma A, Demirovic A, Rao S, Young C, Aas PA, Ericsson I, Sundan A, Jensen ON, Slupphaug G. Modulation of cell metabolic pathways and oxidative stress signaling contribute to acquired melphalan resistance in multiple myeloma cells. PLoS One. 2015;10(3):e0119857. doi: 10.1371/journal.pone.0119857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, Chen Q, Gindin M, Gubin MM, van der Windt GJ, Tonc E, Schreiber RD, Pearce EJ, Pearce EL. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–41. doi: 10.1016/j.cell.2015.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB. The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002;16(6):769–77. doi: 10.1016/s1074-7613(02)00323-0. [DOI] [PubMed] [Google Scholar]
- 130.Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG, Rathmell JC. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011;186(6):3299–303. doi: 10.4049/jimmunol.1003613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Huang SC, Everts B, Ivanova Y, O’Sullivan D, Nascimento M, Smith AM, Beatty W, Love-Gregory L, Lam WY, O’Neill CM, Yan C, Du H, Abumrad NA, Urban JF, Jr, Artyomov MN, Pearce EL, Pearce EJ. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat Immunol. 2014;15(9):846–55. doi: 10.1038/ni.2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ding ZC, Zhou G. Cytotoxic chemotherapy and CD4+ effector T cells: an emerging alliance for durable antitumor effects. Clin Dev Immunol. 2012;2012:890178. doi: 10.1155/2012/890178. [DOI] [PMC free article] [PubMed] [Google Scholar]
