Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jul 1.
Published in final edited form as: Cancer J. 2010 Jul–Aug;16(4):295–303. doi: 10.1097/PPO.0b013e3181eb5066

Chemoimmunotherapy

Leisha A Emens 1
PMCID: PMC2919833  NIHMSID: NIHMS221701  PMID: 20693839

Abstract

Cancer chemotherapy drugs are historically regarded as detrimental to immunity due to their myelosuppressive effects. However, accumulating data suggest that the antitumor activity of conventional cancer chemotherapy results in part from its ability to harness the innate and adaptive immune systems by inducing immunologically active tumor cell death. Additional data broaden the immunologic impact of cancer chemotherapy drugs, demonstrating that some drugs have the ability to disrupt pathways of immune suppression and immune tolerance in a manner that depends on the drug dose, and the timing of its administration in relation to immunotherapy. Understanding the cellular and molecular basis of the interactions between chemotherapy drugs and the immune system will facilitate the strategic development of chemoimmunotherapy treatment regimens that both maximize tumor regression and the antitumor immune response for the long term clinical benefit of cancer patients.

Keywords: immunotherapy, chemotherapy, chemoimmunotherapy, cancer vaccines, adoptive cellular therapy, clinical trials

Introduction

Cancer therapy has evolved to strategically integrate distinct treatment modalities in order to optimize the chance of cure. Surgery and radiation therapy are used to achieve locoregional control, whereas systemic therapies (chemotherapy, endocrine therapy, molecularly targeted therapies, and adjunctive therapies (bisphosphonates)) are used to control diffuse disease (in hematologic malignancies) or disease that has spread beyond the primary site (in solid tumors). Multiple drugs with complementary mechanisms of action and non-overlapping toxicities are frequently combined for additive or synergistic antitumor efficacy. Cancer chemotherapy drugs typically disrupt fundamental regulatory pathways essential for tumor cell growth and survival, but are too often limited by both primary and acquired mechanisms of drug resistance and collateral toxic damage to normal host tissues. In addition, their use historically fails to consider the potential impact of systemic therapies on the overall tumor microenvironment established by the host-tumor interaction. These fundamental limitations to the traditional use of chemotherapy for systemic disease control calls for a radical but strategic shift in the way tumors are treated in order to achieve better clinical outcomes.

More recently, the host-tumor interaction has definitively emerged as a major determinant of clinical course and treatment outcomes for human malignancies13. Elements of the host response to cellular transformation critical for tumor growth and progression include stromal fibroblasts, host endothelial cells, tumor-associated macrophages (TAMs), natural killer (NK) cells, and tumor-specific lymphocytes. Multiple drugs in common use manipulate the host-tumor interaction to favor tumor regression. Bevacizumab, sunitinib, and sorafenib are used to target endothelial cell biology and disrupt the tumor-associated vasculature4. Multiple monoclonal antibodies uniquely marry the specificity of tumor cell-targeted therapy and immunomodulation by specifically binding to tumor cells, then recruiting host innate immune effectors (NK cells and monocytes) to mediate tumor cell cytotoxicity5. Cancer vaccines can actively induce tumor-specific immune responses by eliciting both a humoral (antibody) and a cellular (T cell) immune response6. Advantages of the therapeutic antigen-specific host immune response compared to traditional cancer treatments include exquisite tumor cell specificity, minimal off-target side effects, and the potential for a durable, lasting treatment effect in the absence of continuous, active therapy due to immunologic memory. The first personalized therapeutic cancer vaccine, sipuleucel-T (Provenge), was recently approved by the US Food and Drug Administration for the treatment of advanced prostate cancer. The approval of a drug specifically designed to induce a therapeutic tumor-specific immune response marks the beginning of an era of an entirely different approach to cancer therapy.

Active immune-based approaches to cancer therapy will clearly add a new dimension to cancer care, but they are unlikely to replace traditional cancer therapies. Moreover, the complex interactions between cancer cells and host elements within the tumor microenvironment imply that targeting one aspect of tumor biology will have clear consequences for other elements involved in tumor growth and progression. Thus, chemotherapy not only kills cancer cells directly, but also impairs endothelial cells and impacts tumor immunity7. Anti-angiogenic therapies not only impact tumor-associated endothelial cells, but also modulate tumor immunity8. Tumor-specific monoclonal antibodies not only kill cancer cells directly, but also recruit immune effectors to the therapeutic effect9. Thoughtful consideration of the impact of established cancer drugs like chemotherapy on the vaccines designed to elicit an antitumor immune response (or adoptive cellular therapy) will be required for the most effective integration of immune-based therapy into multimodality cancer care. Current data suggest that combination chemoimmunotherapy regimens have great potential for optimizing the clinical outcomes of cancer patients.

Tumor Immunity and Chemotherapy

Pre-existing mechanisms of immune tolerance and immune escape establish a harsh landscape for effective immune-mediated tumor rejection10. This includes deletion of high avidity tumor-specific T cells, the accumulation of CD4+CD25+FoxP3+ regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs), the secretion of transforming growth factor-β (TGF-β), tumor necrosis factor (TNF), and/or interleukin 10 (IL-10)), the expression of negative co-stimulatory molecules by tumor cells, and mechanisms of immune escape (evolution of antigen loss variants and/or MHC-negative tumor cells, or tumor cells with defects in antigen processing). Compounding this, many standard and high dose chemotherapy regimens are immunosuppressive, inducing or contributing to lymphopenia. Some of these regimens require the co-administration of glucocorticoids, which can be both directly lympholytic and immunosuppressive11. In addition, cancer chemotherapy frequently results in cell death by apoptosis rather than necrosis1214. Apoptosis is a program of cell death long thought to be immunologically inert15, which would further decrease the likelihood of effective immune-mediated tumor destruction.

In contrast, a growing body of evidence suggests that chemotherapy may augment tumor immunity by a variety of mechanisms (Table 1). Very recent evidence suggests that apoptosis, or at least a subset of it, can be highly immunogenic16. Supporting this concept, standard neoadjuvant therapy has been associated with the development of immune cell infiltrates17. Chemotherapy-induced cell death by apoptosis can enhance cross-priming, thereby increasing the antitumor T cell response13. Alternatively, chemotherapy can be used in distinct ways specifically for its immunomodulatory potential18. First, chemotherapy can condition the tumor microenvironment by modulating the expression of tumor antigens, accessory molecules of T cell activation or inhibition, and molecules involved in antigen processing and presentation. Second, chemotherapy can be used to manipulate systemic pathways of immune tolerance and regulation. These direct immune-modulating effects of chemotherapy are not only drug-dependent, but also dependent on drug dose and schedule18. For example, a single low dose of cyclophosphamide given 1–3 days prior to antigen exposure can overcome immune tolerance, augmenting both humoral and cellular immunity. Conversely, cyclophosphamide given concurrently with or subsequent to an antigen exposure induces immune tolerance.

Table 1.

The Immunomodulatory Effects of Chemotherapy Drugs

Chemotherapy Drug Immunologic Process Mechanism
Doxorubicin
Daunorubicicn
Mitoxantrone
Antigen uptake Cell surface calreticulin
Doxorubicin
Daunorubicicn
Mitoxantrone
Oxaliplatin
Antigen processing HMGB1 release
High dose chemotherapy Homeostatic proliferation Cytokines (IL-7, IL-15, IL-21); inhibition of
Treg
Cyclophosphamide
Cisplatin
Paclitaxel
Temozolomide
Fludaribine
Gemcitabine/FOLFOX4
Regulatory cell function Inhibition of CD4+CD25+ FoxP3+ regulatory T
cells
Gemcitabine
5-fluorouracil
Regulatory cell function Reduced MDSC numbers
Cyclophosphamide
Cyclophosphamide/Doxorubicin
Regulatory cell function Increased MDSC numbers
Cyclophosphamide
Paclitaxel
Bleomycin
Melphalan
Immunologic skew Promotes T helper type 1 immunity
Cyclophosphamide
Vincristine
Vinblastine
Paclitaxel
Methotrexate
Mitomycin-C
Doxorubicin
5-aza-2’-deoxycytidine
Antigen presentation Augment DC function
   Cyclophosphamide: decreased CD8+ DC
   Paclitaxel: through TLR-4/MyD88
5-aza-2’-deoxycytidine Antigen processing Increasing MHC Class I expression
5-aza-2’-deoxycytidine
5-Fluorouracil
Antigen processing Increasing tumor antigen expression
Melphalan
Mitomycin-C
Cytosine arabinoside
Co-stimulation Upregulate B7-1/B7-2 expression
Cytosine arabinoside Counter-stimulation Downregulate B7-H1 expression
Cyclophosphamide
Paclitaxel
Cisplatin
Doxorubicin
Target cell lysis Upregulation of lytic sensitivity to TRAIL, fas,
granzyme-B

Abbreviations: HMGB-1=high mobility box globulin-1; IL=interleukin; MDSC=myeloid-derived suppressor cells; DC=dendritic cells; TLR=toll-like receptor; MHC=major histocompatibility complex

Mechanisms of Chemotherapy-Induced Immunomodulation

Immunogenic Cell Death

Many chemotherapy drugs exert their therapeutic effect by the induction of apoptosis, or programmed cell death. This may occur through the enhancement of pre-existing immunity by treatment-related apoptosis. This mechanism has been demonstrated in murine models with gemcitabine, doxorubicin, cyclophosphamide, and paclitaxel in vivo13,1923 At least one study has suggested the relevance of this mechanism in humans. The neoadjuvant therapy of locally advanced breast cancer with paclitaxel resulted in the new accumulation of tumor infiltrating lymphocytes post-treatment, where the extent of T cell infiltration correlated with clinical response (0% with stable disease, 25% with partial clinical response, and 67% with complete clinical response but some residual pathologic disease)17. The extent of tumor cell apoptosis with the first paclitaxel treatment predicted the accumulation of TIL and clinical benefit.

For some drugs, including anthracyclines (doxorubicin, daunorubicin, and mitoxantrone) and oxaliplatin, the mechanism underlying immunogenic tumor cell death post-chemotherapy has been elucidated12,17,24. The observation that some drugs induce apoptosis that is immunogenic and others induce apoptosis that is not suggests that alternate pathways of apoptosis may exist16. Studies have shown that one major difference between immunogenic and nonimmunogenic apoptosis is the expression of calreticulin (CRT) on the cell surface16. While normally expressed only in the endoplasmic reticulum, CRT undergoes caspase-1-dependent translocation to the cell surface, representing one of the earliest markers of cellular stress preceding cell death. Cell surface expression of CRT greatly enhances the uptake of dying tumor cells by dendritic cells (DC). Importantly, CRT is necessary, but not sufficient, for the immune response induced by apoptosis. The alarmin high mobility globulin box-1 (HMGB1) is released by cells dying in response to immunogenic chemotherapy. HMGB1 (a non-histone chromatin-binding protein that regulates gene transcription and nuclear functions when present within the nucleus25) binds to the toll-like receptor 4 (TLR-4), triggering DC maturation and the activation of antigen-specific CD4+ and CD8+ T cell immunity12. Thus, CRT is essential for engulfment and subsequent DC maturation, whereas the HMGB1-TLR-4 interaction is required for optimal processing and presentation of tumor antigens from the dying tumor cells to T lymphocytes12. The clinical relevance of this pathway is revealed by a TLR-4 polymorphism leading to a single amino acid substitution (asp299gly) in the TLR-4 extracellular domain. This polymorphism reduces the binding of HMGB1 to human TLR-4 and inhibits HMGB1 mediated DC-T cell interactions. DCs from patients with a TLR-4 asp299gly mutation do not cross-present antigens from dying melanoma cells to CD8+ T cells in vitro12. Moreoever, TLR-4 asp299gly mutation carriers, accounting for about 17% of a breast cancer cohort, had a higher risk of disease relapse after adjuvant treatment with anthracycline-based chemotherapy12. Similar findings have been reported for advanced colon cancer patients treated with oxaliplatin24.

A second danger signal released by tumor cells dying in response to immunogenic chemotherapy is adenosine triphosphate (ATP)26,27. ATP binds to the P2RX7 purinergic receptor on DC, thereby stimulating assembly of the NOD-like receptor family, pyrin-domain-containing protein-3 (NRLP3)-dependent caspase-1 activation complex—the NRLP3 inflammasome--thus resulting in the proteolytic maturation of caspase-1, and the cleavage of pro-interleukin-1β to release soluble interleukin-1β (IL-1β). IL-1β then promotes the priming of interferon-γ (IFN-γ)-producing, tumor-specific CD8+ T cells. Validating the clinical relevance of this pathway is the observation that breast cancer patients with a loss of function allele of P2RX7 relapsed sooner after anthracycline-based chemotherapy than breast cancer patients with a normal P2RX7 allele26,27. Other danger signals include crystalline uric acid (UA), heat shock proteins (hsp) 70 and 90, and the NK cell ligand NKG2D28.

Homeostatic Proliferation

While some evidence suggests that lymphopenia may be associated with worse clinical outcomes29, data also suggest that profound lymphopenia can create an environment for “re-booting” the immune system30,31. If profound lymphopenia is established therapeutically (for example, with chemotherapy), a period of enhanced T cell proliferation follows, driven by cytokines that include interleukin-7, interleukin-15, and interleukin-213. This proliferation is driven by the recognition of self antigens, presented in the context of MHC Class I molecules. Thus, the tolerance for altered self characteristic of human tumors can be disrupted by homeostatic proliferation. This can be further skewed toward effective tumor immunity by the adoptive transfer of lymphocytes or vaccination during homeostatic T cell proliferation, which directs the re-established T cell repertoire toward a desired antigenic specificity32. Vaccine-induced tumor immunity can be enhanced when tumor-bearing mice are vaccinated with granulocyte-macrophage colony-stimulating (GM-CSF)-secreting tumor vaccines during early engraftment after syngeneic or allogeneic T cell-depleted bone marrow transplantation33,34, and further enhanced by donor leukocyte infusion from vaccinated donor mice35. Furthermore, breast 4T1 tumor-bearing mice that undergo surgical resection of existing tumor, followed by nonmyeloablative allogeneic stem cell transplantation and donor leukocyte infusions plus vaccination with a GM-CSF-secreting tumor vaccine developed immune responses capable of lysing metastatic 4T1 breast tumors36. These concepts have been tested in clinical trials of adoptive T cell transfer alone or combined with immunization during immune reconstitution37,38. Significant numbers of tumor antigen-specific T lymphocytes were shown to be present in individuals that displayed tumor regression after adoptive cellular therapy. Optimizing host conditioning, and characterizing the kinetics, longevity, and functional quality of tumor antigen-specific T cells after immune reconstitution is essential for the optimal application of this approach to cancer care.

Regulatory T Cells (Treg)

It is now clear that a distinct population of suppressive T cells plays a key role in maintaining peripheral immune tolerance. CD4+CD25+ regulatory T cells (Treg) represent 5–10% of the peripheral T lymphocyte pool in mice and humans, and exist both to downregulate the normal immune response and prevent autoimmunity39. They express CTLA-4, GITR, and FoxP3, and secrete IL-10 and TGF-β. Treg inhibit CD8+ T cell responses in an interleukin-2 (IL-2)-dependent manner through either direct cell-cell contact or the immunosuppressive effects of IL-10 or TGF-β. Treg potently suppress antitumor immune responses and accumulate in the peripheral blood and tumor microenvironment of patients with cancers of the breast, pancreas, ovary, stomach, lung, and liver40.

Multiple groups have shown in preclinical models that treatment with low dose cyclophosphamide mitigates the influence of CD4+CD25+ Treg, allowing tumor immunity to emerge4144. Cyclophosphamide reverses tumor-induced immunologic skew, promoting CD4+ T helper type 1-driven tumor immunity45,46. Administering a low dose of cyclophosphamide prior to vaccination can facilitate the recruitment of latent, high aviditiy CD8+ T cells that mediate tumor rejection41. Pretreatment of mice bearing HPV E7+ tumors with cisplatin can decrease Treg and enhance antigen-specific CD8+ T cell activity in response to E7-targeted DNA vaccination47. The activity of Treg can also be diminished in mice by giving cyclophosphamide, paclitaxel, or temozolamide in metronomic fashion4850.

The immune-modulating impact of chemotherapy drugs has also been explored in cnacer patients. Metronomic cyclophosphamide depletes CD4+C25+ Treg and restores T and NK effector function in patients with late stage cancers51. Standard paclitaxel-based chemotherapy in non small cell lung cancer (NSCLC) patients selectively decreased Treg through fas-mediated apoptosis, and upregulated the T helper type 1 cytokines IFN-γ and IL-2, and CD44 in CD4+ and CD8+ effector T cells52. Standard dose fludarabine can also decrease the number and function of Treg in patients with B cell chronic lymphocytic leukemia53. In addition, treatment of patients with metastatic colorectal carcinoma with gemcitabine and FOLFOX4, followed by subcutaneous GM-CSF and IL-2, produces a significant reduction of Treg in about 65% of patients; this finding is associated with a 70% objective response rate to therapy54,55.

Myeloid-Derived Suppressor Cells (MDSC)

Myeloid-derived suppressor cells (MDSC) are a diverse population of cells that consists of myeloid progenitor cells and immature myeloid cells56. Murine MDSC are GR1+CD11b+, whereas human MDSC are typically CD14CD11b+CD33+HLADR. These cells expand in both tumor-bearing mice and cancer patients (up to 10-fold), and markedly suppress T cell-mediated immune responses through increased nitric oxide (NO) and arginase (ARG) production56.

Chemotherapy can also influence MDSC. Cyclophosphamide causes a spike a MDSC numbers in tumor-free mice57, and MDSC numbers increase in breast cancer patients treated with dose dense doxorubicin-cyclophosphamide (AC)58. Conversely, standard dose gemcitabine can eliminate MDSC in mice, thereby enhancing the activity of CD8+ T cells and NK cells59. Cisplatin given prior to vaccination can decrease levels of peripheral MDSC in tumor-bearing mice47.

Dendritic Cells

Dendritic cells (DC) are specialized antigen presenting cells that both induce self tolerance and stimulate T cell activation60. Conventional (as opposed to plasmacytoid) DC includes tissue-derived migratory DC and lymphoid tissue-resident DC. Migratory DC translocate from peripheral tissues to locoregional lymph nodes and are restricted to these lymph nodes, whereas resident DC are replenished by bone marrow-derived precursors61. Resident DC can be CD8+, CD4+, or CD4CD860. Migratory DC can transfer antigen to resident DC, although the relative contribution of these two subsets to immunologic homeostasis remains unclear60,62.

A number of chemotherapy drugs given at low doses have been reported to augment dendritic cells function, including cyclophosphamide, vincristine, vinblastine, paclitaxel, methotrexate, mitomycin-C, doxorubicin, and 5-aza-2’-deoxycytidine63,64. Of these drugs, the impact of cyclophosphamide and paclitaxel on DC maturation and function has been best described. Low dose cyclophosphamide selectively decreases CD8+ resident DC compared to migratory or plasmacytoid DC, resulting in enhanced antigen presentation, augmented cytokine secretion, and some inhibition of Treg; this effect is abrogated by the adoptive transfer of CD8+ DC to cyclophosphamide-treated mice, confirming the cell type modulated65. Extending this observation, nonmyeloablative doses of cyclophosphamide induce a rebound myelopoiesis with ensuing trafficking of DC to tumors, where they secrete more IL-12 and less IL-10 to favorably expand effector T cells over Treg66. Treatment with low dose cyclophosphamide has been associated with increased expression of DC maturation markers67, and also regulates the production of type 1 interferons, thereby promoting the evolution of the CD44hi memory T cell response68. Highlighting the dose-dependent immunomodulatory activity of cyclophosphamide, lymphodepleting doses of cyclophosphamide induce the proliferation of DC in the bone marrow, followed by their expansion in the periphery69. These dendritic cells are capable of antigen presentation to T cells in vivo70. Notably, although lymphodepleting doses of cyclophopshamide also induce the systemic expansion of MDSC, these cells neither differentiate into DC nor impact their expansion or function57.

The taxanes also have immune-modulating effects on DC. Paclitaxel has lipopolysaccharide (LPS)-mimetic activity, interacting through the murine toll-like receptor 4 (TLR-4) to facilitate DC activation and the secretion of pro-inflammatory cytokines71,72. A similar Myd88-dependent but TLR-4 independent effect suggests that an alternative TLR is responsible for the paclitaxel effect in humans73. Several preclinical studies have shown that paclitaxel can augment the activity of tumor vaccines, augmenting antigen-specific immunity and tumor-free survival23,46,74,75; this has been demonstrated for docetaxel as well76,77. Subclinical doses of paclitaxel given one day prior to (but not one week after) a HER-2-targeted GM-CSF-secreting cell-based vaccine augment CD4+ T cell-dependent immunity in tolerant neu-N mice, delaying tumor outgrowth46. Interestingly, combining low dose paclitaxel with low dose cyclophosphamide synergize to delay tumor outgrowth in this preclinical model78. Paclitaxel-exposed DC precursors typically display higher levels of costimulatory molecules, maturation markers, IL-12 production, and augmented CD8+ T cell priming and cytotoxic activity in vitro and in vivo78,79.

Chemomodulation of Tumor Cell Immunogenicity

In addition to inducing apoptosis and immunologic cell death, some chemotherapy drugs render tumor cells more immunogenic in other ways. Chemotherapy can modulate the expression of both tumor antigens and molecules that regulate antigen processing and presentation. 5-fluorouracil can enhance the expression of carcinoembryonic antigen (CEA) in colon and breast carcinoma cells80, and 5’-aza-2’deoxycytidine8184 can induce the expression of a variety of cancer testis antigens and/or upregulate cell surface MHC Class I expression in distinct tumor cell lines (renal cell carcinoma, ovarian carcinoma, glioma, and melanoma), rendering them more susceptible to antigen-specific CD8+ T cell-mediated lysis. Melphalan and mitomycin-C can upregulate expression of costimulatory B7 molecules, thus enabling tumor cells themselves to present antigen with a concurrent costimulatory signal85,86. Cytosine arabinoside (ara-C) increases the expression of B7-1 and B7-2 and decreases the expression of B7-H1, thus enhancing CD8+ T cell-mediated killing in murine models of acute myelogenous leukemia (AML)87. This effect may underlie the ability of ara-C combined with a GM-CSF-secreting tumor cell vaccine to prolong the survival of AML-bearing mice88. Supporting the relevance of these observations for humans, a similar impact of ara-C on the expression of costimulatory molecules was observed for a majority of primary cultured human AML cells in vitro87. Multiple chemotherapeutic drugs can sensitize tumor cells to CTL-mediated apoptosis through fas- or perforin-granzyme-mediated pathways in vitro89. Furthermore, a single dose of cyclophosphamide can cure mice of malignant mesothelioma by sensitizing the mesothlioma cells to TRAIL-dependent CD8+ T cell-mediated apoptosis even in the absence of effective antigen-specific T cell expansion90.

More recently, the treatment of mice bearing Lewis lung carcinoma cells with low dose paclitaxel combined with an intratumoral DC vaccine was associated with enhanced numbers of tumor infiltrating CD4+ and CD8+ T cells, and elevated tumor-specific IFN-γ production by draining lymph node cells91. In vitro, paclitaxel pretreated tumor cells did not inhibit DC maturation to the extent that untreated tumor cells did. Furthermore, low dose paclitaxel altered the cytokine profile within the tumor site in vivo, with increased monocyte chemotactic protein-1 (MCP-1) and chemokine (C-X-C motif) ligand-10 (IP-10) levels and decreased levels of interleukin-1α (IL-1α). Finally, paclitaxel, cisplatin, and doxorubicin can sensitize tumor cells to T cell-mediated lysis by making them permeable to granzyme B92. This effect is perforin-independent, and mediated by the upregulation of mannose-6-phosphate in tumor cells. It was associated also with bystander lysis of tumor cells that did not express the tumor antigen target.

Clinical Trials of Chemoimmunotherapy

Clinical evidence suggests that the human immune system can be impacted by standard dose chemotherapy in both negative and positive ways (Table 2). Studies have shown that vaccination in close proximity to standard dose cytotoxic chemotherapy can inhibit vaccine-induced immune responses, facilitate levels of immune priming similar to those in the absence of chemotherapy, or increase the response to subsequent chemotherapy. One study of examined CEA-specific immune responses after 60 patients with advanced, CEA-expressing colorectal cancer were vaccinated with the canary pox vaccine ALVAC-CEA-B7.193. The number of CEA-specific T cells was decreased in patients who received a greater number of prior chemotherapy regimens and in those who had most recently received standard dose chemotherapy. Another study vaccinated patients with Stage II and III pancreatic carcinoma with a GM-CSF-secreting cell-based vaccine once after primary pancreaticoduodenectomy, and documented the induction of mesothelin-specific T cells post-vaccination94,95. Patients were then treated with 6 months of 5-fluorouracil-based chemoradiation, after which the mesothelin response was undetectable. Vaccine-induced mesothelin-specific immunity was restored only after three additional vaccines were given after chemoradiation was complete, suggesting suppression of the vaccine-induced immune response by subsequent chemoradiation. Based on promising evidence of immunologic activity in Phase I and II clinical trial of vaccine alone96, a recent Phase III clinical trial (VITAL-2) of a GM-CSF-secreting, cell-based prostate cancer vaccine enrolled 405 chemotherapy-naïve men with symptomatic, hormone-refractory prostate cancer97. Subjects were randomized to receive vaccination with 5×108 cell prime followed by 3×108 cell boost every 2 weeks for 12 cycles plus docetaxel 75 mg/m2 every 3 weeks for 10 cycles, followed by 3×108 cell boost every 4 weeks, or docetaxel 75 mg/m2 every 3 weeks plus daily prednisone 10 mg daily for 10 cycles. Halabi predicted survival time was 13 months for both arms. In August 2008, the Independent Data Monitoring Committee (IDMC) noted an imbalance of deaths between the two arms, and recommended study termination. The imbalance then lessened from 20 to 9, with 85 and 75 deaths on the two arms. All deaths were due to disease progression and death from prostate cancer. Importantly, the combination of this vaccine and standard dose docetaxel had not been tested in early Phase I and II clinical trials. Thus, the failure of this Phase III trial may be due in part due to a failure to appreciate the impact of standard dose docetaxel therapy on vaccine-induced tumor immunity.

Table 2.

Clinical Trials of Combinatorial Chemoimmunotherapy

Standard Dose Chemotherapy
Patient Population n Vaccine Drug Immunologic Outcome
metastatic colon cancer 17 CEA peptide standard 5-fluorouracil,
leucovorin, irinotecan
CEA-specific immunity in 50% of
vaccinated patients
metastatic colon cancer 118 ALVAC-CEAB7.1 standard 5-fluorouracil,
leucovorin, irinotecan
no inhibition of CEA-specific
immunity
early stage pancreas cancer 60 GM-CSF-secreting
mesothelin+ pancreas
tumor cells
vaccine once, then 5-
fluorouracil-based chemo-
radiation, then 3 additional
vaccines
Inhibiton of vaccine-induced
mesothelin T cell responses by
subsequent chemoradiation,
restored by 3 additional vaccines
metastatic prostate cancer 28 PSA vaccinia/fowl pox standard docetaxel and
dexamethasone
no inhibition of PSA-specific
immunity
metastatic melanoma 10 Melan-A/gp100 peptide DTIC 0 mg/m2 (n=5) vs.
DTIC 800 mg/m2 (n=5)
increased peptide-specific
immunity with DTIC 800 mg/m2
metastatic hormone-refractory
prostate cancer
405 GM-CSF-secreting
prostate tumor cells
Vaccine plus docetaxel vs.
docetaxel plus prednisone
NR, study terminated
Low Dose Immunomodulatory Chemotherapy
Patient Population n Vaccine Drug Immunologic Outcome
metastatic breast cancer 1028 KLH (n=505)
KLH-STn (n=523)
CY 300 mg/m2 NR, no clinical difference
metastatic pancreas cancer 50 GM-CSF-secreting
mesothelin+ pancreas
tumor cells
CY 0 mg/m2 (n=30)
CY 300 mg/m2 (n=20)
increased mesothelin-specific
immunity with CY 300 mg/m2
metastatic breast cancer 28 GM-CSF-secreting
HER-2+ breast tumor
cells
CY 0, 200, 250, 350 mg/m2
DOX 0, 15, 25, 35 mg/m2
(factorial design of various
dose combinations)
increased HER-2-specific
immunity with CY 200 mg/m2
plus DOX 35 mg/m2

Abbreviations: CEA=carcinoembryonic antigen; PSA=prostate-specific antigen; DTIC=dacarbazine; KLH=keyhole limpet hemocyanin; STn=clustered carbohydrate antigens; CY=cyclophosphamide; NR=not reported; GM-CSF=granulocyte-macrophage colony-stimulating factor; DOX=doxorubicin

On the other hand, standard chemotherapy can interact with established vaccine-induced immune responses for clinical benefit. A platform of hematopoietic cell transplantation after high dose chemotherapy with or without radiation followed by either adoptive cellular therapy or vaccine therapy has been tested by multiple groups. One study of cellular therapy transferred T cells specific for minor histocompatibility antigens after allogeneic stem cell transplantation for recurrent leukemia in 7 patients98. The primary toxicity was pulmonary. Transferred T cells persisted for up to 21 days, and 5 of the 7 patients achieved complete but transient remissions post-therapy, thus demonstrating the feasibility of this approach. Another report tested adoptive cellular therapy with autologous tumor-infiltrating lymphocytes (TIL) and IL-2 in 50 patients with metastatic melanoma equally randomized to receive conditioning with cyclophosphamide plus fludarabine with either 2 or 12 Gray of total body radiation (TBI)99. Nonmyeloablative chemotherapy alone resulted in response rates of 49%, whereas the addition of 2 or 12 Gray of TBI gave response rates of 52% and 72% respectively. Lymphodepletion was associated with elevated serum levels of IL-7 and IL-15, and objective responses correlated with the telomere length of the transferred lymphocytes. The use of GM-CSF-secreting, cell-based autologous vaccines after autologous stem cell transplantation for AML was evaluated in two distinct trials, with both demonstrating evidence of safety and immunogenicity100,101.

Other groups have tested the impact of standard dose chemotherapy sequenced prior to or after vaccine therapy for solid tumors. A recent report of 36 HLA-A2+ disease-free, stage II-IV melanoma patients tested standard dose dacarbazine (DTIC) at 800 mg/m2 given one day prior to vaccination with melan-A/MART-1 plus gp100 melanoma peptide vaccination compared to vaccination alone102. DTIC enhanced the numbers of peptide-specific CD8+ effector T cells, and the generation and persistence of peptide-specific effector memory CD8+ T cells induced by peptide vaccination (Table 2). In another study, twenty-nine patients with extensive stage small cell lung cancer (SCLC) were vaccinated with DC transduced with an adenoviral vector expressing wild-type 53 (DC-Adp53)103. Although almost 60% of patients developed p53-specific immune responses with vaccine alone, all but one developed progressive disease. Interestingly, of those that received subsequent salvage chemotherapy, those that did develop p53-specific immunity were much more likely to display an objective clinical response. This observation suggests that previous vaccination may sensitize patients to respond to subsequent chemotherapy, and additional trials are studying the impact of previous therapeutic cancer vaccination with the DC-Adp53 vaccine on the subsequent response to chemotherapy in a prospective fashion104. A clinical trial of 32 patients with glioblastoma vaccinated with an autologous DC vaccine also demonstrated the apparent sensitization of patients with a vaccine-induced immune response to subsequent chemotherapy, with improved time to progression spanning post-vaccination salvage chemotherapy but not improved time to progression spanning the time period post-vaccination alone in patients who developed immunity105. A recent preclinical study using the adoptive transfer of nonspecifically activated CD4+ T cells as a chemosensitizer before the administration of chemotherapy demonstrated a marked enhancement of the cytotoxic effect of chemotherapeutic drugs both in vitro and in vivo, providing some preclinical experimental validation for these clinical observations106.

Work has also examined the impact of concurrent chemotherapy on cancer vaccines (Table 2). A study of prime-boost vaccination alone or with concurrent docetaxel therapy enrolled 28 patients with metastatic, hormone refractory prostate cancer107. The vaccination regimen consisted of a priming immunization with a recombinant vaccinia virus expressing prostate specific antigen (PSA) admixed with recombinant vaccinia virus expressing B7.1 followed by recombinant fowlpox virus expressing PSA. Vaccination resulted in over a 3-fold increase in PSA-specific T cells by ELISPOT at 3 months, regardless of concurrent docetaxel. Median progression-free survival for vaccinated patients on docetaxel was 6.1 months compared to 3.7 months for a historical cohort of patients treated with docetaxel alone. A prospective study to follow up on observations that prior chemotherapy may inhibit cancer vaccine activity randomized 118 patients with metastatic colorectal cancer to receive ALVAC alone for 3 vaccination cycles followed by ALVAC given with chemotherapy (fluorouracil/leucovorin/irinotecan), ALVAC with tetanus toxoid adjuvant for three vaccination cycles followed by ALVAC given with chemotherapy, or 4 cycles of chemotherapy followed by 4 cycles of ALVAC vaccination in patients without disease progression108. Increases in CEA-specific T cells were detected in 50%, 37%, and 30% of patients respectively, suggesting that systemic chemotherapy did not impact vaccine-induced immune responses. Another study tested the combination of three cycles of standard irinotecan/high dose 5-fluorouracil/leucovorin and concurrent vaccination with a carcinoembryonic antigen (CEA)-derived peptide, followed by vaccination with peptide alone in patients with newly diagnosed metastatic colorectal cancer109. Almost half of treated patients developed CEA-specific T cell responses by intracellular cytokine staining.

Several clinical trials have now been reported that use low, immune-modulating doses of chemotherapy specifically to enhance the activity of cancer vaccines rather than to lyse tumor cells directly (Table 2). A number of Phase II studies designed to mitigate the influence of suppressor T cell as they were defined some 35 years ago first showed that patients receiving cyclophosphamide at 300 mg/m2 three days before vaccination with a clustered carbohydrate antigen (STn)-keyhole limpet hemocyanin (KLH) vaccine developed higher antibody titers and enjoyed longer survival110. In follow up to these early studies, a Phase III clinical trial of 1,028 women with metastataic breast cancer randomized 505 women to receive cyclophosphamide plus KLH and 523 women to receive cyclophosphamide plus STn-KLH111. In this trial, no difference in time to disease progression or overall survival emerged. This approach has also been tested in combination with GM-CSF-secreting tumor vaccines for a variety of cancer. One trial enrolled patients with metastatic pancreatic cancer, testing immune-modulating doses of cyclophosphamide (300 mg/m2) given one day prior to immunization with a GM-CSF-secreting, cell-based pancreas cancer vaccine112. This study revealed a trend toward increased vaccine-activated CD8+ T cell immunity specific for mesothelin, and a corresponding trend toward increased clinical benefit with cyclophosphamide-modulated vaccination compared to vaccination alone. Similar findings regarding clinical benefit were reported in small trial of the same vaccine platform in patients with NSCLC treated with immune-modulating doses of cyclophosphamide at 300 mg/m2 given the day prior to vaccination; a transient decrement in Treg numbers with time after cyclophosphamide treatment was also observed 113. Another clinical trial with an innovative factorial response surface design tested a HER-2-positive, GM-CSF-secreting cell-based breast tumor vaccine alone or with a range of low doses of cyclophosphamide (0, 200, 250, 350 mg/m2) given one day prior to vaccination and doxorubicin (0, 15, 25, 35 mg/m2) given seven days after vaccination in patients with stable metastatic breast cancer114. The vaccine alone induced de novo HER-2-specific delayed type hypersensitivity (DTH), with low levels of HER-2-specific antibody also induced. The addition of cyclophosphamide at 200 mg/m2 maintained the DTH response and further augmented HER-2-specific antibody levels, but cyclophosphamide doses of 250 mg/m2 or higher abrogated vaccine-induced immunity. The chemotherapy dose combination that optimized vaccine activity was 200 mg/m2 cyclophosphamide, and 35 mg/m2 doxorubicin. It is notable that most clinical cancer vaccine trials have historically used cyclophosphamide at doses of 250–300 mg/m2 for immune-modulation to enhance vaccine activity, suggesting a narrow therapeutic window for the immunomodulatory activity of low dose cyclophosphamide.

Conclusions

We have made a great deal of progress in understanding the cellular and molecular basis of both the fundamental immunobiology of the host-tumor interaction, and the impact of cancer chemotherapy drugs on that interaction. Armed with this knowledge, the potential for maximizing the bioactivity and clinical benefit of immune-based therapies has never been greater. Strategically integrating immunotherapies with chemotherapy drugs in order to shape the overall host milieu and the local tumor microenvironment and ameliorate distinct mechanisms of immune tolerance and suppression will ultimately support a vigorous and sustained antitumor immune response. Carefully dissecting the proper dose and timing of drugs for integrating with immune-based therapies in clinically relevant laboratory models and early phase clinical trials will accelerate late stage clinical development, making clinically meaningful chemoimmunotherapy for cancer care a reality.

Acknowledgements

This work was supported by funding from the Department of Defense (W81XWH-04-1-0595; W81XWH-07-1-0485), the Breast SPORE program (P50 CA88843), the National Institutes of Health (K23 CA098498-01), the American Cancer Society (RSG CCE-112685), Komen for the Cure (BCTR0707297), the AVON Foundation, the Gateway Foundation for Cancer Research, the V Foundation for Cancer Research, and Climb for Hope.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest Statement

This review describes work using granulocyte-macrophage colony-stimulating factor-secreting tumor vaccines. Under a licensing agreement between Biosante, Incorporated and the Johns Hopkins University, the University is entitled to a share of royalty received by the University on sales of products described in this article. The terms of this agreement are being managed by the Johns Hopkins University in accordance with its conflict of interest policies.

References

  • 1.Formenti S. Immunological aspects of local radiotherapy: clinical relevance. Discov Med. 2010;9:119–1224. [PubMed] [Google Scholar]
  • 2.Pages F, Galon J, Dieu-Nosjean M, et al. Immune infiltration in human tumors: a prognostic factor that should not be ignored. Oncogene. 2010;29:1093–1102. doi: 10.1038/onc.2009.416. [DOI] [PubMed] [Google Scholar]
  • 3.Zitvogel L, Apetoh L, Ghiringhelli F, et al. Immunological aspects of cancer chemotherapy. Nat Rev Immunol. 2008;8:59–73. doi: 10.1038/nri2216. [DOI] [PubMed] [Google Scholar]
  • 4.Heath V, Bicknell R. Anticancer strategies involving the vasculature. Nat Rev Clin Oncol. 2009:395–404. doi: 10.1038/nrclinonc.2009.52. [DOI] [PubMed] [Google Scholar]
  • 5.Weiner L, Dhodapkar M, Ferrone S. Monoclonal antibodies for cancer immunotherapy. Lancet. 2009;373:1033–1040. doi: 10.1016/S0140-6736(09)60251-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Finn O. Cancer immunology. N Engl J Med. 2008;358:2704–2715. doi: 10.1056/NEJMra072739. [DOI] [PubMed] [Google Scholar]
  • 7.Emens L. Chemotherapy and tumor immunity: an unexpected collaboration. Front Bio. 2008:249–257. doi: 10.2741/2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Johnson B, Osada T, Clay, et al. Physiology and therapeutics of vascular endothelial growth factor in tumor immunosuppression. Curr Mol Med. 2009;9:702–707. doi: 10.2174/156652409788970634. [DOI] [PubMed] [Google Scholar]
  • 9.Weiner L, Surana R, Wang S. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nat Rev Immunol. 2010;10:317–327. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Emens L, Jaffee E. Immunotherapy and cancer therapeutics: why partner? In: Prendergast GC, Jaffee EM, editors. Cancer Immunotherapy: Immune Suppression and Tumor Growth. London: Academic Press, Elsevier; 2007. pp. 207–233. [Google Scholar]
  • 11.Herold M, McPherson K, Reichardt H. Glucocorticoids in T cell apoptosis and function. Cell Mol Life Sci. 2006;63:60–72. doi: 10.1007/s00018-005-5390-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat Med. 2007;13:1050–1059. doi: 10.1038/nm1622. [DOI] [PubMed] [Google Scholar]
  • 13.Casares N, Pequignot N, Tesniere A, et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J Exp Med. 2006;202:1691–1701. doi: 10.1084/jem.20050915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Obeid M, Tesniere A, Ghiringhelli F, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007;13:54–61. doi: 10.1038/nm1523. [DOI] [PubMed] [Google Scholar]
  • 15.Green D, Ferguson T, Zitvogel L, et al. Immunogenic and tolerogenic cell death. Nat Rev Immunol. 2009;9:353–363. doi: 10.1038/nri2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zitvogel L, Kepp O, Senovilla L, et al. Immunogenic tumor cell death for optimal anticancer therapy: the calreticulin exposure pathway. Clin Cancer Res. 2010 doi: 10.1158/1078-0432.CCR-09-2891. 10.1158/1078/0432.CCR-09-2891. [DOI] [PubMed] [Google Scholar]
  • 17.Demaria S, Volm M, Shapiro R, et al. Development of tumor-infiltrating lymphocytes in breast cancer after neoadjuvant paclitaxel chemotherapy. Clin Cancer Res. 2001;7:3025–3303. [PubMed] [Google Scholar]
  • 18.Emens L, Machiels J, Reilly R, et al. Chemotherapy: friend or foe to cancer vaccines? Curr Opin Mol Ther. 2001;3:77–84. [PubMed] [Google Scholar]
  • 19.Nowak A, Lake R, Marzo A, et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. J Immunol. 2003;170:4905–4913. doi: 10.4049/jimmunol.170.10.4905. [DOI] [PubMed] [Google Scholar]
  • 20.Nowak A, Robinson B, Lake R. Gemcitabine exerts a selective effect on the humoral immune response: implications for combination chemo-immunotherapy. Cancer Res. 2002;62:2353–2358. [PubMed] [Google Scholar]
  • 21.Nowak A, Robinson B, Lake R. Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res. 2003;63:4490–4496. [PubMed] [Google Scholar]
  • 22.Tong Y, Song W, Crystal R. Combined intratumoral injection of bone-marrow-derived dendritic cells and systemic chemotherapy to treat pre-existing murine tumors. Cancer Res. 2001;61:7530–7535. [PubMed] [Google Scholar]
  • 23.Yu B, Kusmartsev S, Cheng F, et al. Effective combination of chemotherapy and dendritic cell administration for the treatment of advanced stage experimental breast cancer. Clin Cancer Res. 2003;9:285–294. [PubMed] [Google Scholar]
  • 24.Tesniere A, Schlemmer F, Boige V, et al. Immunogenic death of colon cancer cells treated with oxaliplatin. Oncogene. 2009 doi: 10.1038/onc.2009.356. epub ahead of print PMID: 19881547. [DOI] [PubMed] [Google Scholar]
  • 25.Lotze M, Tracey K. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol. 2005;5:331–342. doi: 10.1038/nri1594. [DOI] [PubMed] [Google Scholar]
  • 26.Aymeric L, Apetoh L, Ghiringhelli F, et al. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 2010;70:855–858. doi: 10.1158/0008-5472.CAN-09-3566. [DOI] [PubMed] [Google Scholar]
  • 27.Ghiringhelli F, Apetoh L, Tesniere A, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL1β-dependent adaptive immunity against tumors. Nat Med. 2009;15:1170–1179. doi: 10.1038/nm.2028. [DOI] [PubMed] [Google Scholar]
  • 28.Locher C, Rusakiewicz S, Tesniere A, et al. Witch hunt against tumor cells enhanced by dendritic cells. Ann NY Acad Sci. 2009;1174:51–60. doi: 10.1111/j.1749-6632.2009.04940.x. [DOI] [PubMed] [Google Scholar]
  • 29.Lissoni P, Brivio F, Fumagalli L, et al. Efficacy of cancer chemotherapy in relation to the pretreatment number of lymphocytes in patients with metastatic solid tumors. Int J Biol Markers. 2004;19:135–140. doi: 10.1177/172460080401900208. [DOI] [PubMed] [Google Scholar]
  • 30.Cho B, Rao V, Ge Q, et al. Homeostasis-stimulated proliferation drives naive T cells to differentiate directly into memory T cells. J Exp Med. 2000;192:549–556. doi: 10.1084/jem.192.4.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Goldrath A, Bogatzki L, Bevan M. Naive T cells transiently aquire a memory-like phenotype during homeostasis-driven proliferation. J Exp Med. 2000;192:557–564. doi: 10.1084/jem.192.4.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mackall C, Bare C, Granger L, et al. Thymic-independent T cell regeneration occurs via antigen-driven expansion of peripheral T cells resulting in a repertoire that is limited in diversity and prone to skewing. J Immunol. 1996;156:4609–4616. [PubMed] [Google Scholar]
  • 33.Borrello I, Sotomayor E, Rattis F-M, et al. Sustaining the graft-versus-tumor effect through posttransplant immunization with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing tumor vaccines. Blood. 2000;95:3011–3019. [PubMed] [Google Scholar]
  • 34.Teshima T, Mach N, Hill G, et al. Tumor cell vaccine elicits potent antitumor immunity after allogeneic T-cell-depleted bone marrow transplantation. Cancer Res. 2001;61:162–171. [PubMed] [Google Scholar]
  • 35.Teshima T, Liu C, Lowler K, et al. Donor leukocyte infusion from immunized donors increases tumor vaccine efficacy after allogeneic bone marrow transplantation. Cancer Res. 2002;62:796–800. [PubMed] [Google Scholar]
  • 36.Luznik L, Slansky J, Jalla S, et al. Successful therapy of metastatic cancer using tumor vaccines in mixed allogeneic bone marrow chimeras. Blood. 2003;101:1645–1652. doi: 10.1182/blood-2002-07-2233. [DOI] [PubMed] [Google Scholar]
  • 37.Dudley M, Wunderlich J, Robbins P, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science. 2002;298:850–854. doi: 10.1126/science.1076514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dudley M, Wunderlich J, Yang J, et al. Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastastic melanoma. J Clin Oncol. 2005;23:2346–2357. doi: 10.1200/JCO.2005.00.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zou W. Regulatory T cells, tumor immunity, and immunotherapy. Nat Rev Immunol. 2006;6:295–307. doi: 10.1038/nri1806. [DOI] [PubMed] [Google Scholar]
  • 40.Nizar S, Copier J, Meyer B, et al. T-regulatory cell modulation: the future of cancer immunotherapy. Br J Cancer. 2009;100:1697–1703. doi: 10.1038/sj.bjc.6605040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ercolini A, Ladle B, Manning E, et al. Recruitment of latent pools of high-avidity CD8+ T cells to the antitumor immune response. J Exp Med. 2005;201:1591–1602. doi: 10.1084/jem.20042167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4+CD25+ regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol. 2004;34:336–344. doi: 10.1002/eji.200324181. [DOI] [PubMed] [Google Scholar]
  • 43.Lutsiak M, Semnani R, De Pascalis R, et al. Inhibition of CD4+CD25+ T regulatory cell function implicated in enhanced immune response by low dose cyclophosphamide. Blood. 2005;105:2862–2868. doi: 10.1182/blood-2004-06-2410. [DOI] [PubMed] [Google Scholar]
  • 44.Taieb J, Chaput N, Schartz N, et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. J Immunol. 2006;176:2722–2729. doi: 10.4049/jimmunol.176.5.2722. [DOI] [PubMed] [Google Scholar]
  • 45.Ding Z, Blazar B, Mellor A, et al. Chemotherapy rescues tumor-driven aberrant CD4+ T cell differentiation and restores an activated polyfunctional helper phenotype. Blood. 2010;115:2397–2406. doi: 10.1182/blood-2009-11-253336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Machiels J, Reilly R, Emens L, et al. Cyclophosphamide, doxorubicin, and paclitaxel enhance the antitumor immune response of granulocyte/macrophage-colony stimulating factor-secreting whole-cell vaccines in HER-2/neu tolerized mice. Cancer Res. 2001;61:3689–3697. [PubMed] [Google Scholar]
  • 47.Tseng C-W, Hung C-F, Alvarez R, et al. Pretreatment with cisplatin enhances E7-specific CD8+ T cell-mediated antitumor immunity induced by DNA vaccination. Clin Cancer Res. 2008;14:3185–3192. doi: 10.1158/1078-0432.CCR-08-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Banissi C, Ghiringhelli F, Carpentier A. Treg depletion with a low-dose metronomic temozolomide regimen in a rat glioma model. Cancer Immunol Immunother. 2009;58:1627–1634. doi: 10.1007/s00262-009-0671-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chen C-A, Ho C-M, Chang M-C, et al. Metronomic chemotherapy enhances antitumor effects of cancer vaccine by depleting regulatory T lymphocytes and inhibiting tumor angiogenesis. Mol Ther. 2010 doi: 10.1038/mt.2010.34. doi:10.1038/mt.2010.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hermans I, Chong T, Palmowski M, et al. Synergistic effect of metronomic dosing of cyclophosphamide combined with specific antitumor immunotherapy in a murine melanoma model. Cancer Res. 2003;63:8408–8413. [PubMed] [Google Scholar]
  • 51.Ghiringhelli F, Menard C, Puig P, et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector function in end stage cancer patients. Cancer Immunol Immunother. 2007;56:641–648. doi: 10.1007/s00262-006-0225-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang L, Dermawan K, Jin M, et al. Differential impairment of regulatory T cells rather than effector T cells by paclitaxel-based chemotherapy. Clin Immunol. 2008;129:219–229. doi: 10.1016/j.clim.2008.07.013. [DOI] [PubMed] [Google Scholar]
  • 53.Beyer M, Kochanek M, Darabi K, et al. Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine. Blood. 2005;106:2018–2025. doi: 10.1182/blood-2005-02-0642. [DOI] [PubMed] [Google Scholar]
  • 54.Correale P, Cusi M, Tsang K, et al. Chemo-immunotherapy of metastatic colorectal carcinoma with gemcitabine plus FOLFOX 4 followed by subcutaneous granulocyte macrophage colony stimulating factor and interleukin-2 induces strong immunologic and antitumor activity in metastatic colon cancer patients. J Clin Oncol. 2005;23:8950–8958. doi: 10.1200/JCO.2005.12.147. [DOI] [PubMed] [Google Scholar]
  • 55.Correale P, Tagliaferri P, Fioravanti A, et al. Immunity feedback and clinical outcome in colon cancer patients undergoing chemoimmunotherapy with gemcitabine+FOLFOX followed by subcutaneous granulocyte macrophage colony-stimulating factor and aldesleukin (GOLFIG-1. Clin Cancer Res. 2008;14:4192–4199. doi: 10.1158/1078-0432.CCR-07-5278. [DOI] [PubMed] [Google Scholar]
  • 56.Gabrilovich D, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162–174. doi: 10.1038/nri2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Salem M, Al-Khami A, El-Naggar S, et al. Cyclophosphamide induces dynamic alterations in the host microenvironments resulting in a Flt3 ligand-dependent expansion of dendritic cells. J Immunol. 2010;184:1737–1747. doi: 10.4049/jimmunol.0902309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Diaz-Montero C, Salem M, Nishimura M, et al. Increased circulating myeloid-derived suppressor cells correlated with clinical cancer stage, metastatic tumor burden, and doxorubicin-cyclophosphamide chemotherapy. Cancer Immunol Immunother. 2008;58:49–59. doi: 10.1007/s00262-008-0523-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Le H, Graham L, Cha E, et al. Gemcitabine directly inhibits myeloid-derived suppressor cells in BALB/c mice bearing 4T1 mammary carcinoma and augments expansion of T cells from tumor-bearing mice. Int Immunopharm. 2009;9:900–909. doi: 10.1016/j.intimp.2009.03.015. [DOI] [PubMed] [Google Scholar]
  • 60.Villadangos J, Schnorrer P. Intrinsic and cooperative antigen-presenting functions of dendritic cell subsets in vivo. Nat Rev Immunol. 2007;7:543–555. doi: 10.1038/nri2103. [DOI] [PubMed] [Google Scholar]
  • 61.Lui K, Waskow C, Liu X, et al. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat Immunol. 2007;8:578–583. doi: 10.1038/ni1462. [DOI] [PubMed] [Google Scholar]
  • 62.Allenspach E, Lemos M, Porrett P, et al. Migratory and lymphoid-resident dendritic cells cooperate to efficiently prime naive CD4+ T cells. Immunity. 2008;29:795–806. doi: 10.1016/j.immuni.2008.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kaneno R, Shurin G, Tourkova I, et al. Chemomodulation of human dendritic cell function by antineoplastic agents in low noncytotoxic concentrations. J Transl Med. 2009;7:58. doi: 10.1186/1479-5876-7-58. DOI 10.1186/1479-5876-7-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Shurin G, Tourkova I, Kaneno R, et al. Chemotherapeutic agents in noncytotoxic concentrations increase antigen presentation by dendritic cells via an IL12-dependent mechanism. J Immunol. 2009;183:137–144. doi: 10.4049/jimmunol.0900734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nakahara T, Uchi H, Lesokhin A, et al. Cyclophosphamide enhances immunity by modulating the balance of dendritic cell subsets in lymphoid organs. Blood. 2010 doi: 10.1182/blood-2009-11-251231. DOI 10.1182/blood-2009-11-251231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Radojvic V, Bezak K, Skarica M, et al. Cyclophosphamide resets dendritic cell homeostasis and enhances antitumor immunity through effects that extend beyond regulatory T cell elimination. Cancer Immunol Immunother. 2010;59:137–148. doi: 10.1007/s00262-009-0734-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wada S, Yoshimura K, Hipkiss E, et al. Cyclophosphamide augments antitumor immunity: studies in an autochthonous prostate cancer model. Cancer Res. 2009;69:4309–4318. doi: 10.1158/0008-5472.CAN-08-4102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Schiavoni G, Mattei F, Di Puchio T, et al. Cyclophosphamide induces type I interferon and augments the number of CD44high T lymphocytes in mice: implications for strategies of chemoimmunotherapy of cancer. Blood. 2000;95:2024–2030. [PubMed] [Google Scholar]
  • 69.Salem M, Diaz-Montero C, Al-Khami A, et al. Recovery from cyclophosphamide-induced lymphopenia results in expansion of immature dendritic cells which can mediate enhanced prime-boost vaccination antitumor responses in vivo when stimulated with the TLR3 agonist poly(I:C) J Immunol. 2009;182:2030–2040. doi: 10.4049/jimmunol.0801829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Salem M, El-Naggar S, Cole D. Cyclophosphamide induces bone marrow to yield higher numbers of precursor dendritic cells in vitro capable of functional antigen presentation to T cells in vivo. Cell Immunol. 2010;261:134–143. doi: 10.1016/j.cellimm.2009.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Byrd-Leifer C, Block E, Takeda K, et al. The role of MyD88 and TLR4 in the LPS-mimetic activity of Taxol. Eur J Immunol. 2001;31:2448–2457. doi: 10.1002/1521-4141(200108)31:8<2448::aid-immu2448>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 72.Kawasaki K, Akashi S, Shimazu R, et al. Mouse toll-like receptor 4.MD-2 complex mediates lipopolysaccharide-mimetic signal transduction by Taxol. J Biol Chem. 2000;275:2251–2254. doi: 10.1074/jbc.275.4.2251. [DOI] [PubMed] [Google Scholar]
  • 73.Wang J, Kobayashi M, Han M, et al. MyD88 is involved in the signaling pathway for Taxol-induced apoptosis and TNF-alpha expression in human myelomonocytic cells. Br J Hematol. 2002;11:638–645. doi: 10.1046/j.1365-2141.2002.03645.x. [DOI] [PubMed] [Google Scholar]
  • 74.Chopra A, Kim T, O-Sullivan I, et al. Combined therapy of an established, highly aggressive breast cancer in mice with paclitaxel and a unique DNA-based vaccine. Int J Cancer. 2006;118:2888–2898. doi: 10.1002/ijc.21724. [DOI] [PubMed] [Google Scholar]
  • 75.Eralp Y, Wang X, Wang J, et al. Doxorubicin and paclitaxel enhance the antitumor efficacy of vaccines directed against HER-2/neu in a murine mammary carcinoma model. Breast Cancer Res. 2004;6:R275–R283. doi: 10.1186/bcr787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chu Y, Wang L, Yang G, et al. Efficacy of GM-CSF-producing tumor vaccine after docetaxel chemotherapy in mice bearing established Lewis lung carcinomas. J Immunother. 2006;29:367–380. doi: 10.1097/01.cji.0000199198.43587.ba. [DOI] [PubMed] [Google Scholar]
  • 77.Prell R, Gearin L, Simmons A, et al. The anti-tumor efficacy of a GM-CSF-secreting tumor cell vaccine is not inhibited by docetaxel administration. Cancer Immunol Immunother. 2006;12:1–9. doi: 10.1007/s00262-005-0116-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Pfannenstiel L, Lam S, Emens L, et al. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol. 2010;263:79–87. doi: 10.1016/j.cellimm.2010.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.John J, Ismail M, Riley C, et al. Differential effects of paclitaxel on dendritic cell function. BMC Immunol. 2010;19:14–24. doi: 10.1186/1471-2172-11-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Correale P, Aquino A, Giuliani A, et al. Treatment of colon and breast carcinoma cells with 5-fluorouracil enhances expression of carcinoembryonic antigen and susceptibility to HLA-A*02/01 restricted, CEA-peptide-specific cytotoxic T cells in vitro. Int J Cancer. 2003;104:437–445. doi: 10.1002/ijc.10969. [DOI] [PubMed] [Google Scholar]
  • 81.Adair S, Hogan K. Treatment of ovarian cancer cell lines with 5-aza-2'-deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded molecules. Cancer Immunol Immunother. 2009;58:589–601. doi: 10.1007/s00262-008-0582-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Coral S, Sigalotti L, Altomonte M, et al. 5-aza-2'-Deoxycytidine-induced expression of functional cancer testis antigens in human renal cell carcinoma: immunotherapeutic implications. Clin Cancer Res. 2002;8:2690–2695. [PubMed] [Google Scholar]
  • 83.Fonsatti E, Nicolay H, Sigalotti L, et al. Functional up-regulation of human leukocyte antigen class I antigens expression by 5-aza-2'-deoxycytidine in cutaneous melnaoma: Immunotherapeutic implications. Clin Cancer Res. 2007;13:3333–3338. doi: 10.1158/1078-0432.CCR-06-3091. [DOI] [PubMed] [Google Scholar]
  • 84.Natsume A, Wakabayashi T, Tsujimura K, et al. The DNA methylating agent 5-aza-2'-deoxycytidine activates NY-ESO-1 antigenicity in orthotopic human glioma. Int J Cancer. 2008;122:2542–2553. doi: 10.1002/ijc.23407. [DOI] [PubMed] [Google Scholar]
  • 85.Donepudi M, Raychaudhuri P, Bluestone J, et al. Mechanism of melphalan-induced B7-1 gene expression in P815 tumor cells. J Immunol. 2001;166:6491–6499. doi: 10.4049/jimmunol.166.11.6491. [DOI] [PubMed] [Google Scholar]
  • 86.Sojka D, Donepudi M, Bluestone J, et al. Melphalan and other anticancer modalities up-regulate B7-1 gene expression in tumor cells. J Immunol. 2000;164:6230–6236. doi: 10.4049/jimmunol.164.12.6230. [DOI] [PubMed] [Google Scholar]
  • 87.Vereecque R, Saudemont A, Quesnel B. Cytosine arabinoside induces costimulatory molecule expression in acute myeloid leukemia cells. Leukemia. 2004;18:1223–1230. doi: 10.1038/sj.leu.2403391. [DOI] [PubMed] [Google Scholar]
  • 88.Lin J, Li B, Rimmer E, et al. Enhancement of the anti-tumor efficacy of GM-CSF-secreting tumor cell immunotherapy in preclinical models by cytosine arabinoside. Exp Hem. 2008;36:319–328. doi: 10.1016/j.exphem.2007.11.005. [DOI] [PubMed] [Google Scholar]
  • 89.Yang F, Haluska F. Treatment of melanoma with 5-fluorouracil or dacarbazine in vitro sensitizes cells to antigen-specific CTL lysis through perforin/granzyme- and Fas-mediated pathways. J Immunol. 2004;172:4599–4608. doi: 10.4049/jimmunol.172.7.4599. [DOI] [PubMed] [Google Scholar]
  • 90.van der Most R, Curie A, Cleaver A, et al. Cyclophosphamide chemotherapy sensitizes tumor cells to TRAIL-dependent CD8+ T cell-mediated immune attack resulting in suppression of tumor growth. PLoS One. 2009;4:e6982. doi: 10.1371/journal.pone.0006982. doi:6910.1371/journal.pone.0006982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhong H, Han B, Tourkova I, et al. Low-dose paclitaxel prior to intratumoral dendritic cell vaccine modulates intratumoral cytokine network and lung cancer growth. Clin Cancer Res. 2007;13:5455–5462. doi: 10.1158/1078-0432.CCR-07-0517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ramakrishnan R, Assudani D, Nagaraj S, et al. Chemotherapy enhances tumor cell susceptibility to CTL-mediated killing during cancer immunotherapy in mice. J Clin Invest. 2010;120:1111–1124. doi: 10.1172/JCI40269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.von Mehren M, Arlen P, Gulley J, et al. The influence of granulocyte macrophage colony-stimulating factor and prior chemotherapy on the immunological response to a vaccine (ALVAC-CEA B7.1) in patients with metastatic carcinoma. Clin Cancer Res. 2001;7:1181–1191. [PubMed] [Google Scholar]
  • 94.Jaffee E, Hruban R, Biedrzycki B, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Oncol. 2001;19:145–156. doi: 10.1200/JCO.2001.19.1.145. [DOI] [PubMed] [Google Scholar]
  • 95.Thomas A, Santarsiero L, Lutz E, et al. Mesothelin-specific CD8+ T cell responses provide evidence of in vivo cross-priming by antigen-presenting cells in vaccinated pancreatic cancer patients. J Exp Med. 2004;200:297–306. doi: 10.1084/jem.20031435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Emens L. GM-CSF-secreting vaccines for solid tumors. Curr Op Invest Drugs. 2009;10:1315–1324. [PubMed] [Google Scholar]
  • 97.Small E, Demkow T, Gerritson W, et al. A Phase III trial of GVAX immunotherapy for prostate cancer in combination with docetaxel vs. docetaxel plus prednisone in symptomatic, castration-resistant prostate cancer (CRPC) GU ASCO. 2009 [Google Scholar]
  • 98.Warren E, Fujii N, Akatsuka Y, et al. Therapy of relapsed leukemia after allogeneic hematopoietic cell transplantation with T cells specific for minor histocompatibility antigens. Blood. 2010;115:3869–3878. doi: 10.1182/blood-2009-10-248997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Dudley M, Yang J, Sherry R, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–5239. doi: 10.1200/JCO.2008.16.5449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Borrello I, Levitsky H, Stock W, et al. Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting cellular immunotherapy in combination with autologous stem cell transplantation (ASCT) as postremission therapy for acute myeloid leukemia (AML) Blood. 2009;114:1736–1745. doi: 10.1182/blood-2009-02-205278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ho V, Vanneman M, Kim H, et al. Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation. Proc Natl Acad Sci USA. 2009;106:15825–15830. doi: 10.1073/pnas.0908358106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Nistico P, Capone I, Palermo B, et al. Chemotherapy enhances vaccine-induced antitumor immunity in melanoma patients. Int J Cancer. 2009;124:130–139. doi: 10.1002/ijc.23886. [DOI] [PubMed] [Google Scholar]
  • 103.Antonia S, Mirza N, Fricke I, et al. Combination of p53 cancer vaccine with chemotherapy in patients with extensive stage small cell lung cancer. Clin Cancer Res. 2006;12:878–887. doi: 10.1158/1078-0432.CCR-05-2013. [DOI] [PubMed] [Google Scholar]
  • 104.Chiappori A, Soliman H, Janssen W, et al. INGN-225: a dendritic cell-based p53 vaccine (Ad.p53-DC) in small cell lung cancer: observed associations between immune response and enhanced chemotherapeutic effect. Exp Op Biol Ther. 2010;10:983–991. doi: 10.1517/14712598.2010.484801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wheeler C, Black K, Liu G, et al. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res. 2008;68:5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
  • 106.Radfar S, Wang Y, Khong H. Activated CD4+ T cells dramatically enhance chemotherapeutic tumor responses in vitro and in vivo. J Immunol. 2009;183:6800–6807. doi: 10.4049/jimmunol.0901747. [DOI] [PubMed] [Google Scholar]
  • 107.Arlen P, Gulley JPC, Skarupa L, et al. A randomized phase II study of concurrent docetaxel plus vaccine versus vaccine alone in metastatic androgen-independent prostate cancer. Clin Cancer Res. 2006;12:1260–1269. doi: 10.1158/1078-0432.CCR-05-2059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kaufman H, Lenz H-J, Marshall J, et al. Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colon cancer. Clin Cancer Res. 2008;14:4843–4849. doi: 10.1158/1078-0432.CCR-08-0276. [DOI] [PubMed] [Google Scholar]
  • 109.Weihrauch M, Ansen S, Jurkiewicz E, et al. Phase I/II combined chemoimmunotherapy with carcinoembryonic antigen-derived HLA-A2-restricted CAP-1 peptide and irinotecan, 5-fluorouracil, and leucovorin in patients with primary metastatic colorectal cancer. Clin Cancer Res. 2005;15:5993–6001. doi: 10.1158/1078-0432.CCR-05-0018. [DOI] [PubMed] [Google Scholar]
  • 110.Emens L, Jaffee E. Toward a breast cancer vaccine: work in progress. Oncology (Williston Park) 2003;17:1200–1211. discussion 1214, 1217-120. [PubMed] [Google Scholar]
  • 111.Mayordomo J, Tres A, Miles D, et al. Long-term follow-up of patients concomitantly treated with hormone therapy in a prospective controlled randomized multicenter clinical study comparing STn-KLH vaccine with KLH control in stage IV breast cancer following first line chemotherapy. Proc Am Soc Clin Oncol abstr. 2004;23:188. [Google Scholar]
  • 112.Laheru D, Lutz E, Burke J, et al. Allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor immunotherapy alone or in sequence with cyclophosphamide for metastatic pancreatic cancer: a pilot study of safety, feasibility, and immune activation. Clin Cancer Res. 2008;14:1455–1463. doi: 10.1158/1078-0432.CCR-07-0371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Schiller J, Nemunaitis J, Ross H. A phase 2 randomized study of GM-CSF gene-modified autologous tumor vaccine (CG8123) with and without low dose Cyclophosphamide in advanced stage non-small cell lung cancer (NSCLC). Presented at the Int Assoc for Study Lung Cancer.2005. [Google Scholar]
  • 114.Emens L, Asquith J, Leatherman J, et al. Timed sequential treatment with Cyclophosphamide, Doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: A chemotherapy dose-ranging factorial study of safety and immune activation. J Clin Oncol. 2009;27:5911–5918. doi: 10.1200/JCO.2009.23.3494. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES