Topic Highlight Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Dec 14, 2013; 19(46): 8531-8542
Published online Dec 14, 2013. doi: 10.3748/wjg.v19.i46.8531
Immunotherapy for colorectal cancer
Shigeo Koido, Toshifumi Ohkusa, Kazuki Takakura, Keisuke Saito, Zensho Ito, Hiroko Kobayashi, Mikio Kajihara, Kan Uchiyama, Seiji Arihiro, Hisao Tajiri, Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Chiba 277-8567, Japan
Shigeo Koido, Sadamu Homma, Department of Oncology, Institute of DNA Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
Masato Okamoto, Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan
Shigeo Koido, Yoshihisa Namiki, Institute of Clinical Medicine and Research, The Jikei University School of Medicine, Chiba 277-8567, Japan
Hiroshi Arakawa, Department of Endoscopy, The Jikei University School of Medicine, Chiba 277-8567, Japan
Jianlin Gong, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, United States
Author contributions: Koido S, Ohkusa T, Homma S, Namiki Y, Takakura K, Saito K, Ito Z, Kobayashi H, Kajihara M, Uchiyama K, Arihiro S, Arakawa H, Okamoto M, Gong J and Tajiri H contributed equally to the manuscript, drafting the article and revising it critically for important intellectual content, and approved the final version for publication.
Supported by Grants in Aid for Scientific Research (C) from the Japanese Ministry of Education, Culture, Sports, Science and Technology
Correspondence to: Shigeo Koido, MD, Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, 163-1 Kashiwashita, Kashiwa-shi, Chiba 277-8567, Japan. shigeo_koido@jikei.ac.jp
Telephone: +81-4-71641111 Fax: +81-4-71633488
Received: September 5, 2013
Revised: October 22, 2013
Accepted: November 18, 2013
Published online: December 14, 2013

Abstract

The incidence of colorectal cancer (CRC) is on the rise, and the prognosis for patients with recurrent or metastatic disease is extremely poor. Although chemotherapy and radiation therapy can improve survival rates, it is imperative to integrate alternative strategies such as immunotherapy to improve outcomes for patients with advanced CRC. In this review, we will discuss the effect of immunotherapy for inducing cytotoxic T lymphocytes and the major immunotherapeutic approaches for CRC that are currently in clinical trials, including peptide vaccines, dendritic cell-based cancer vaccines, whole tumor cell vaccines, viral vector-based cancer vaccines, adoptive cell transfer therapy, antibody-based cancer immunotherapy, and cytokine therapy. The possibility of combination therapies will also be discussed along with the challenges presented by tumor escape mechanisms.

Key Words: Colorectal cancer, Cytotoxic T lymphocyte, Dendritic cell, Immunotherapy, Vaccine

Core tip: The prognosis for patients with recurrent or metastatic colorectal cancer (CRC) is extremely poor. Immunotherapy may be effective for treating CRC patients and/or preventing relapse. The immunotherapeutic approaches for CRC, including peptide vaccines, dendritic cell-based cancer vaccines, whole tumor cell vaccines, viral vector-based cancer vaccines, adoptive cell transfer therapy, antibody-based cancer immunotherapy, and cytokine therapy have been demonstrated. The blockade of multiple immune regulatory checkpoints combined with immunotherapy and/or conventional chemotherapy may be effective in treating patients with advanced CRC.



INTRODUCTION

Colorectal cancer (CRC) is the third most common cancer in men (accounting for 10.0% of all cancers) and the second most common cancer in women (accounting for 9.4% of all cancers) worldwide. Additionally, CRC is the fourth most common cause of cancer-related death[1]. Optimization of surgical resection for patients with localized disease has dramatically improved 5 year and 10 year survival rates. The prognosis for patients with resectable tumors depends on the disease stage. CRC patients with distant metastasis have a 12% survival rate[2], and more than 20% of CRC patients have metastatic disease at the time of diagnosis[3,4]. Moreover, despite the fact that 80% of CRC patients with localized disease demonstrate complete macroscopic clearance of the tumor by surgery, 50% of CRC patients will relapse due to the presence of micro-metastasis at the time of surgery[5]. Chemotherapy is approved for the treatment of regionally metastatic CRC, but it shows only modest efficacy and is ineffective against distant metastases[6]. The prognosis for patients with advanced disease remains unfavorable due to the frequency of recurrence, distant metastasis, and resistance to chemotherapy. Thus, novel treatment modalities are needed. Interestingly, tumors that develop chemotherapy or radiation resistance are still suitable targets for immunotherapy[7-10]. Therefore, cancer immunotherapy may be effective for treating CRC patients and/or preventing relapse.

ANTITUMOR IMMUNE RESPONSES
T cells

Tumor cells degrade endogenous and exogenous tumor-associated antigens (TAAs) into short peptides (usually 8-10 amino acids) and present them on the cell surface in the context of major histocompatibility complex (MHC) class I molecules. T cell receptor (TCR) interaction with the complex of peptides and MHC class I molecules on tumor cells is a critical event in the T cell-mediated antitumor immune response. T cells that express the αβ TCR generally express CD4+ (helper T cells) or CD8+ (cytotoxic T cells) lineage markers[11]. In particular, CD8+ T cells recognize peptides (usually 8-10 amino acids) derived from TAAs bound by MHC class I molecules on tumor cells. Thus, immunotherapy may promote cancer cell killing by eliciting antitumor immune responses by recognizing specific TAAs on tumor cells.

To induce antigen-specific CD8+ cytotoxic T lymphocytes (CTLs), peptides derived from TAAs must be presented on the surface of antigen presenting cells (APCs) in the context of MHC class I molecules. In contrast, CD4+ T cells recognize peptides (usually 10-30 amino acids) in association with MHC class II molecules on APCs and enhance the persistence of antigen-specific CD8+ CTLs through secretion of interleukin (IL)-2 and interferon (IFN)-γ[12]. Therefore, the interaction of the αβ TCR with complexes of peptides and MHC class I and class II molecules on APCs is a central event in cancer immunotherapy. The αβ TCR expressed by CD8+ CTLs recognizes MHC class I-peptide complexes on tumor cells and leads to tumor cell killing through effector molecules such as perforin and granzyme B[13]. Moreover, there is increasing evidence that CD4+ T cells play a more direct role in generating efficient antitumor immunity beyond simply assisting[14]. Therefore, effective antitumor responses depend on the presence and function of T cells that recognize and eliminate tumor cells[14,15].

A unique subset of human T cells expresses the TCR-γδ. Human γδT cells include several subsets of cells defined by their TCR. The most common subset of TCR-γδT cells in circulating blood express the Vγ9Vδ2 receptor[16,17]. Although cancer immunotherapy strategies primarily focus on activation of these MHC-restricted T cells, γδT cells and αβT cells share certain effector functions such as cytokine production and potent cytotoxic activity. However, they recognize different sets of antigens, usually in a non-MHC-restricted fashion[16,18]. Thus, T cells can attack tumors in their HLA-unrestricted cytotoxic capacity, as well as by secreting cytokines. Indeed, tumor-infiltrating γδT cells have been detected in a broad range of cancers, including CRC[19]. Importantly, activated γδT cells can kill cells from metastatic renal cell carcinomas, mammary carcinomas, prostate carcinomas and colorectal carcinomas, while sparing normal, untransformed cells[18,19].

Natural killer cells

Natural killer (NK) cells are component of innate immunity responses to tumor cells[20]. NK cells can rapidly kill certain target cells, including tumor cells with down-regulated MHC class I molecules[21]. Thus, NK cells play a critical role in antitumor immunity. NK cells recognize tumor cells via cellular stress or DNA damage signals[22]. Activated NK cells directly kill target tumor cells through several mechanisms, including[23]: (1) cytoplasmic granules such as perforin and granzyme B[24]; (2) tumor necrosis factor-related apoptosis-inducing ligand and Fas ligand (FasL)[25,26]; (3) effector molecules such as IFN-γ and nitric oxide (NO)[24,27]; and (4) antibody-dependent cellular cytotoxicity (ADCC)[28]. NK cell activators (IL-2, IL-12, IL-15, and IL-18), have also been validated in preclinical cancer models[23].

Dendritic cells

Dendritic cells (DCs) are potent APCs that have been used in cancer vaccines due to their ability to initiate antitumor immune responses[12]. DCs are characterized by expression of MHC class I, class II, and costimulatory molecules (B7, ICAM-1, LFA-1, LFA-3, and CD40)[29-31]. These molecules function in concert to generate a network of secondary signals essential for reinforcing the primary antigen-specific signal in T-cell activation[29-31]. DCs process endogenously synthesized antigens into antigenic peptides, which are presented on the cell surface in MHC class I-peptide and recognized by the αβ TCR on naïve CD8+ T cells[12]. DCs can also capture and process exogenous antigens, which are then presented by MHC class I molecules through an endogenous pathway in a process known as “cross-presentation”[32]. Moreover, exogenous antigens from the extracellular environment are also captured by DCs and delivered to the endosomal/lysosomal compartment, where they are degraded to antigenic peptides by proteases and peptidases. These antigens then complex with MHC class II for recognition by the αβ TCR of naïve CD4+ T cells[12]. Efficient antigen presentation by MHC class I and class II on DCs is essential for evoking tumor-specific immune responses[33]. Mature DCs are significantly better at processing and presenting MHC-peptide to the TCR and inducing CTLs due to higher expression of MHC class I and class II and costimulatory molecules[33].

Immature DCs reside in peripheral tissues where they take up and process antigens that are degraded to peptides. These peptides are then bound to MHC class I molecules for presentation to CD8+ CTLs or bound to MHC class II molecules for presentation to CD4+ T helper (Th) cells. Differentiation of the immature DCs into mature DCs is triggered by molecular stimuli that are released in response to tissue disturbance and local inflammatory responses caused by pathogens[34]. After antigen uptake and stimulation by the inflammatory response, immature DCs in the peripheral tissues undergo a maturation process characterized by the up-regulation of MHC class I and class II and costimulatory molecules, the up-regulation of chemokine receptors such as CCR7, and the secretion of cytokines such as IL-12[34,35]. The mature DCs migrate to secondary lymphoid organs, where they present antigens to CD4+ and CD8+ T cells through the MHC class I and class II pathways, respectively[12,34]. Therefore, the aim of immunotherapy is to simultaneously activate CD8+ CTLs (which recognize TAA) and CD4+ Th cells.

Immune suppressive cells

CD4+ Th cells are critical for inducing and regulating immune responses. Immune homeostasis is primarily controlled by two distinct helper T cell subsets, Th1 and Th2 cells[36]. Th1 cells secrete IFN-γ, which can further sensitize tumor cells to CTLs by inducing the up-regulation of MHC class I molecule expression on tumor cells and antigen-processing machinery in DCs[12]. Th2 cells secrete type II cytokines such as IL-4 and IL-10 that enhance humoral immunity (the antibody-based antitumor response)[12]. Importantly, tumor cell-derived soluble factors such as transforming growth factor-β (TGF-β) and IL-10 induce tolerance by promoting the expansion of the CD4+α-2R (CD25)+ forkhead box P3 (Foxp3)+ natural Treg subset[37]. Induced Tregs (CD4+CD25+Foxp3-) secrete TGF-β and IL-10 and suppress Th1 and Th2 phenotypes[38,39]. Therefore, Tregs play a pivotal role in tumor progression and the suppression of antitumor immunity.

The cancer microenvironment consists not only of cancer cells but also stromal cells such as cancer-associated fibroblasts, tolerogenic DCs, myeloid-derived suppressor cells, immunosuppressive tumor-associated macrophages (TAMs), and Tregs. These immune suppressive cells secrete vascular endothelial growth factor (VEGF), IL-6, IL-10, TGF-β, soluble FasL, and indolamine-2,3-dioxygenase (IDO)[40], which inhibit antitumor immunity by various mechanisms, including depletion of arginine and elaboration of reactive oxygen species (ROS) and NO. Moreover, the tumor microenvironment promotes the accumulation of Tregs that suppress CD8+ CTL function due to the secretion of IL-10 or TGF-β from Tregs and tumor cells[40] (Figure 1).

Figure 1
Figure 1 Immunosuppression in the tumor microenvironment. Cancer cells secrete various factors such as vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-10, transforming growth factor-β (TGF-β), Fas ligand (FasL), PD1 ligand 1 (PD-L1), and indolamine-2,3-dioxygenase (IDO), all of which promote the accumulation of heterogeneous populations of cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and tolerogenic dendritic cells (DCs). These immunosuppressive cells inhibit antitumor immunity by various mechanisms, including depletion of arginine and elaboration of reactive oxygen species (ROS) and nitric oxide (NO). The tumor microenvironment promotes the accumulation of Tregs that suppress CD8+ cytotoxic T lymphocyte (CTL) function through secretion of IL-10 and TGF-β.
IMMUNOTHERAPY

Immunotherapy is an active therapeutic approach designed to trigger the immune system to respond to tumor-specific antigens and attack tumor cells. Immunotherapy strategies include the use of peptides derived from TAAs, whole tumor cells, in vitro-generated DCs, or viral vector-based cancer vaccines (Table 1).

Table 1 Immunotherapy strategies for colorectal cancer.
VaccineClinical responseImmunological responseRef.
Peptide
Survivin-2BPR (1/15)SD (3/15) PD (11/15)Temporary decrease of CEA level 40% (6/15)Increase of Survivin-2B-specific CTL frequency DTH 40% (6/15)[48]
Combination chemotherapy with peptide vaccine against RNF43 and TOMM34SD (16/19)PD (3/19)8 of 21 patients exhibited antigen-specific CTL responses to both RNF43 and TOMM34, and 12 patients exhibited CTL responses to one of the peptides only[52]
13-mer mutant rasOf nine patients who completed all six vaccinations, seven patients showed no remaining evidence of diseaseTwo CRC patients showed immunological responses, and the antitumor immune response was significantly associated with prolonged overall survival[53]
β-hCGProlongation of survival in patients with a high level of antipeptide antibodiesInduction of serum antipeptide antibody (56/77)[46]
SART3Diagnosis at 5 mo after first vaccination:SD (1/19) PD (10/19)Increased CTL activity (2/11), induction of serum antipeptide IgG (2/12), IgE (5/12), DTH (0/12)[45]
A set of 10 overlapping p53 synthetic long peptidesInduction of p53-specific CD4+ and CD8+ T-cell responses (9/10), maintained p53-specific CTL reactivity for at least 6 mo (6/9)[50]
DC
DC pulsed with CEA peptide or CEA mRNADisease stabilization was observed in several patientsThe majority of CRC patients demonstrated induction of CEA-specific T cell responses[60-65]
DCs pulsed with CEA-derived altered peptides combined with the adjuvant Flt3 ligand2 of 12 patients exhibited SD for 3 and 6 mo; 2 patients exhibited CR for more than 10 mo; 1 patient had a mixed response with some regression of liver metastasesExpansion of CD8+ T cells that recognize both the native and altered epitopes and possess an effector CTL phenotype[64]
Whole tumor cell
Autologous tumor cells combined with BCGNo significant clinical benefit was seen with whole tumor cell vaccines in surgically resected patients with stage II or III CRCWhen treatment compliance was evaluated, the trend indicated benefits from vaccination in terms of disease-free survival (P = 0.078) and overall survival (P = 0.12)[68]
NDV-infected irradiated autologous tumor cellsA randomized phase III study of 50 patients with resectable CRC liver metastases demonstrated that vaccination with NDV-infected whole tumor cell did not significantly improve overall survival.DTH (21/31)[74,75]
Viral vector
Replication-defective recombinant fowlpox and vaccinia viruses encoding the CEA antigen and TRICOM (B7.1, ICAM-1, and LFA-3)SD (3/9)Induction of CEA-specific CTL (3/9)[79]
Combination chemotherapy and vaccination with a nonreplicating canarypox virus (ALVAC) expressing the CEA and T-cell costimulatory molecule B7.1 (ALVAC-CEA/B7.1)Objective response(42/118)Increases in CEA-specific T cells were detected in patients treated with chemotherapy and booster vaccination[80]
Peptide vaccines

A peptide vaccine is based on the identification and synthesis of epitopes, which can induce TAA-specific antitumor immune responses. CRC cells express TAAs such as carcinoembryonic antigen (CEA)[41,42], mucin 1[41-43], epidermal growth factor receptor (EGFR)[44], squamous cell carcinoma antigen recognized by T cells 3 (SART3)[45], β-human chorionic gonadotropin (β-hCG)[46], Wilms’ Tumor antigen 1 (WT1)[47], Survivin-2B[48], MAGE3[49], p53[50], or mutated KRAS[51], which are potential targets for immunotherapy. Peptide vaccines targeting these TAAs may be a useful approach for immunotherapy in CRC patients.

Peptide vaccines are simple, safe, stable, and economical. Multiple MHC class I-binding peptides have been identified and tested for immunogenicity. Several peptide vaccines for CRC have been tested in phase I clinical trials. Fifteen patients with advanced or recurrent CRC expressing survivin were vaccinated with a peptide derived from HLA-A*2402-restricted epitopes[48]. In 6 patients, tumor marker levels (CEA and CA19-9) decreased transiently during the survivin-2B peptide vaccination. Moreover, in phase I trial of peptide-cocktail vaccines derived from ring finger protein 43 (RNF43) and translocase of the outer mitochondrial membrane 34 (TOMM34), 8 of 21 patients exhibited antigen-specific CTL responses to both RNF43 and TOMM34, and 12 patients exhibited CTL responses to one of the peptides only[52]. The patients who did not demonstrate any CTL responses had the lowest survival rates. By vaccination with a 13-mer mutant ras peptide, 2 of 7 CRC patients showed antitumor immune responses that were significantly associated with prolonged overall survival[53]. Moreover, in a phase II trial, vaccination with the β-hCG peptide induced anti-β-hCG antibody production in 56 of 77 CRC patients[46]. Interestingly, anti-β-hCG antibody induction was associated with longer overall survival[46]. However, some clinical trials report a discrepancy between clinical and immunological responses. In SART3 peptide vaccine therapy, IgE-type anti-peptide antibodies were detected after vaccination; however, immunological responses were not detected in the patients[45]. Peptide vaccines for CRC patients are generally well-tolerated, with no patients requiring cessation due to toxicity; however, a high frequency of reactions were observed at the injection site due to the use of adjuvants such as incomplete Freund’s adjuvant, IL-2, granulocyte-macrophage colony-stimulating factor (GM-CSF), and bacillus Calmette-Guerin (BCG). Importantly, peptide vaccines have shown only limited success in clinical trials. There are several drawbacks to the peptide vaccination strategy, including: (1) limitations due to the patient’s HLA type[54]; (2) ineffectiveness of CD8+ CTLs due to the down-regulation of certain antigens and MHC class I molecules; (3) impaired DC function in patients with advanced cancer[55]; and (4) tumor microenvironments, where immune suppressive cells such as Tregs exist[13]. Synthetic long peptides may be more attractive candidates for peptide-based vaccines. In a phase I/II trial, 10 CRC patients were vaccinated twice with a set of 10 overlapping p53 synthetic long peptides[50]. P53-specific CD4+ and CD8+ T-cell responses were observed in 9 of 10 CRC patients, and 6 of 9 tested patients maintained p53-specific T-cell reactivity for at least 6 mo. New trials may focus on improving the long peptide vaccine-induced antitumor immune responses.

DC vaccines

Three signals were required for induction of efficient CTL responses: (1) simultaneous presentation of multiple TAAs by both MHC class I and class II molecules; (2) costimulation by membrane-bound receptor-ligand pairs; and (3) cytokines to direct polarization of the resultant antitumor immune responses. DCs can provide all three of these signals that are essential for the induction of antitumor immunity[33]. Therefore, many clinical trials of antigen-pulsed DCs have been conducted in patients with various types of tumors, including CRC.

To date, various strategies for delivering TAAs to DCs have been developed to generate potent CTL responses against tumor cells. DCs have been pulsed with synthetic peptides derived from the known TAAs[56], tumor cell lysates[57], apoptotic tumor cells[32], and tumor RNA[58], or physically fused with whole tumor cells[59] to induce efficient antitumor immune responses (Figure 2). Because CEA is a tumor-associated antigen expressed by most CRCs, DCs have been pulsed with CEA peptides[60-64] or CEA mRNA[63,65]. In these phase I clinical trials, the majority of vaccinated CRC patients demonstrated the induction of CEA-specific T cell responses. Moreover, disease progression stabilized in several patients, and the vaccine was safe and well-tolerated. As CEA is a self-antigen and poorly immunogenic, Fong et al[64] generated altered peptide ligands that were derived from native T cell epitopes and contained amino acid substitutions that either increased the peptide affinity for the MHC peptide-binding groove or modified interactions with the T cell receptor. In this trial, 12 patients were immunized with DCs loaded with altered peptides derived from CEA and the Flt3 adjuvant ligand. Two of 12 patients showed disease stabilization for 3 mo and 6 mo, 2 patients showed complete responses for more than 10 mo, and one patient had a mixed response with some regression of liver metastases. To improve the clinical efficacy of DC-based cancer vaccines, it is crucial to design novel strategies that boost adaptive antitumor immunity to overcome tolerance.

Figure 2
Figure 2 Dendritic cell vaccines. Dendritic cells (DCs) are loaded with antigens, which are taken up and degraded into peptide fragments by antigen-presenting cells such as immature DCs. DCs process tumor-derived peptides and major histocompatibility complex (MHC) class I peptides derived from DCs. They form MHC class I-peptide complexes in the endoplasmic reticulum, which are then transported to the surface of the DCs and presented to CD8+ T cells. DCs also synthesize MHC class II peptides in the endoplasmic reticulum, which are transported to the cytoplasm where MHC class II-peptide complexes are assembled with tumor-derived peptides and presented to CD4+ T cells. CD8+ T cells become cytotoxic T lymphocytes (CTLs), which destroy cancer cells through effector molecules such as perforin and granzyme B. TCR: T cell receptor.
Whole tumor cell vaccines

Because autologous tumor cells express a whole TAAs including those known and unidentified, using whole tumor cells could greatly diminish the chance of tumor escape compared to using a single epitope peptide[41,42]. A significant disadvantage to this approach is the difficulty in generating a “universal” vaccine that could be applicable to all patients with a given cancer. Autologous whole tumor cells have been used as cancer vaccines to induce polyclonal CTL induction in several cancer types[66,67], including CRC[68]. A randomized phase III clinical trial of a combined autologous whole tumor cell plus BCG vaccine was conducted to determine whether surgical resection plus vaccination was more beneficial than resection alone in 412 stage II and III CRC patients. This study showed no significant clinical benefit from whole tumor cell vaccination in surgically resected patients with stage II or III CRC. However, effective immune responses were associated with the improved disease-free and overall survival. Only a small proportion of the proteins in an autologous whole tumor cell vaccine are specific to tumor cells, while a vast majority of antigens in the vaccine are shared with normal cells. Moreover, whole tumor cell vaccines are likely to be poorly immunogenic. Therefore, the immune response generated by whole tumor cell vaccines is generally insufficient to provide benefit to patients. To improve the immunogenicity of whole tumor cell vaccines, autologous tumor cells have been genetically modified to secrete GM-CSF and then re-administered to the patient[69]. The trials have shown promising results in patients with prostate carcinoma[70], renal cell carcinoma[71], metastatic non-small-cell lung carcinoma[72], and melanoma[73]. This approach is based on the fact that GM-CSF is required at the site of the tumor to effectively prime TAA-specific immunity[69]. Another tumor cell vaccine approach utilizes Newcastle disease virus (NDV)-infected irradiated tumor cells as autologous CRC vaccines[74]. This approach resulted in a 97.9% two-year survival rate in patients with resected CRC, compared to 66.7% when treated with autologous tumor cells combined with BCG. However, a randomized phase III study of 50 patients with resectable CRC liver metastases demonstrated that vaccination with NDV-infected whole tumor cells did not significantly improve overall survival, disease-free survival or metastases-free survival, although subgroup analyses suggested that the vaccines were somewhat beneficial[75]. The immunogenicity of whole tumor cells needs to be improved for this vaccination strategy to be effective. However, it is unclear which specific agents (such as cytotoxic chemotherapeutics, ionizing irradiation, and chemical agents) are best suited for killing tumor cells to generate highly immunogenic whole tumor cell vaccines.

Viral vector vaccines

Recombinant viral vectors are potentially useful vaccine vehicles for cancer therapy. Many types of recombinant viruses are naturally immunogenic, infect APCs (specifically DCs), and express TAAs[76]. The natural immunogenicity of viral vectors acts as an adjuvant to help boost TAA-specific immune responses. In one study, CRC patients were immunized with vaccinia virus or a replication-defective avian poxvirus encoding CEA. In this phase I study, viral-based vaccination with replication-defective recombinant fowlpox and vaccinia viruses encoding the CEA antigen and TRICOM (B7.1, ICAM-1, and LFA-3) induced CEA-specific T cell responses[77] and disease stabilization in 40% of patients with metastatic cancer (including CRC) for at least 4 mo[78]. A phase II clinical trial in patients with metastatic CRC examined the efficacy of chemotherapy combined with vaccination with a nonreplicating canarypox virus (ALVAC) expressing the CEA and T-cell costimulatory molecule, B7.1 (ALVAC-CEA/B7.1). Anti-CEA-specific T cell responses were successfully generated in 50% of patients undergoing chemotherapy and booster vaccination. Objective clinical responses were observed in 40% of the patients[79,80]. Interestingly, chemotherapy does not appear to inhibit vaccine-mediated immunity.

ADOPTIVE CELL TRANSFER THERAPY

Cancer immunotherapy can be either active or passive. In passive cellular immunotherapy, specific effector cells are directly infused and are not induced or expanded within the cancer patient. Because most tumor cells express MHC class I-peptide, which can be recognized by antigen-specific CD8+ CTLs. Therefore, adoptive transfer of activated CTLs successfully used in patients with advanced cancer[81]. In adoptive cell transfer therapy (ACT), autologous T cells are removed from patients, activated, expanded to large numbers in vitro and transferred back into the patients. ACT overcomes tolerogenic mechanisms by enabling the selection and activation of highly reactive T cell subpopulations and manipulating the host environment into which the T cells are introduced. Indeed, upon the successful induction of specific CTLs in vitro, a clinical response to adoptive immunotherapy can be expected even in patients with advanced CRC[82]. Moreover, injection of IFN promotes the MHC class I-peptide on the cell surface, thus antitumor immune responses are augmented. However, there are several drawbacks to ACT that should be considered, including a potential lack of immune memory, poor persistence of activated effector cells in patients, the prohibitive expense, and the time required to expand the cells.

A new approach using T cells genetically modified to express receptors that recognize TAAs with high specificity to tumor cells may provide significant clinical benefits, especially for large solid tumors[83]. Recently, several clinical trials have demonstrated the therapeutic potential of this approach, which lead to impressive tumor regression in cancer patients[84]. A phase I trial in CRC patients examined human T cells engineered to express a high-avidity CEA-specific TCRs[85]. In this study, autologous T cells genetically engineered to express a murine TCR against human CEA were administered to three patients with metastatic colorectal cancer that was refractory to standard treatments. All patients experienced profound decreases in serum CEA levels (74%-99%), and one patient had an objective regression of cancer metastatic to the lung and liver. However, all three patients developed severe transient inflammatory colitis.

ANTIBODY-BASED CANCER IMMUNOTHERAPY

Monoclonal antibodies (mAbs) that target surface antigens expressed on tumor cells are clinically effective as cancer therapeutics[86]. Three mAbs (Cetuximab, Bevacizumab and Panitumumab) are approved for the treatment of CRC in the United States, and many other mAbs are being tested in clinical trials[87]. Treatment with mAbs can induce tumor cell death by several mechanisms, including interference with vital signaling pathways. Moreover, it is becoming apparent that innate immune effector mechanisms that engage the Fc portion of the antibody via Fc receptors are equally important[88]. The immune cytotoxicity includes ADCC, complement-mediated cytotoxicity, and antibody-dependent cellular phagocytosis. Bevacizumab, a recombinant humanized monoclonal antibody that selectively binds to human VEGF, is effective in KRAS wild-type CRC patients[89]. Recent evidence has also shown clinical benefits from treatment with anti-EGFR, Cetuximab and Panitumumab in KRAS wild-type CRC patients[90].

CYTOKINE THERAPY

Cytokines are substances proteins and glycoproteins that are secreted by immune cells. They have autocrine and paracrine functions and function locally or at a distance to enhance or suppress antitumor immunity. To date, IL-2 and IFN-α are two cytokines approved by the FDA for cancer therapy. Cytokines may be useful for treating hematologic malignancies (hairy cell leukemia and chronic myelogenous leukemia) or immunogenic tumors (melanoma and renal cell carcinoma). The major cytokines currently in use or under evaluation for cancer therapy are IFN-α, IL-2, GM-CSF, and IL-12.

COMBINED IMMUNOTHERAPY

It is well known that even if CRC appears to have been eradicated by chemotherapy and radiation, a small cancer stem cell (CSC) fraction that can self-propagate and sustain tumor growth frequently persists, leading to relapse and therapeutic failure. Although CSC is often resistant to a variety of treatments, including chemotherapy and radiotherapy, immunotherapy may still be effective[8-10]. A combined approach that uses conventional chemotherapy or radiation to kill the bulk of cancer cells and immunotherapy to keep residual CSCs and differentiated cancer cells in check may abrogate the replenishing pool of CRC cells[91]. In addition, treatment with chemotherapy such as cyclophosphamide or gemcitabine can augment the antitumor effects of cancer immunotherapy by depleting Treg, potentially enhancing antitumor immune responses[92]. Therefore, chemotherapy can kill cancer cells and boost antitumor immune responses all at the same time[93,94]. A recent study reported that immune checkpoint blockade with monoclonal antibodies targeting the inhibitory immune receptors cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), PD-1, and PD-L1 can be used to successfully treat patients with advanced melanoma (Figure 3)[95-98]. Combined, these approaches have the potential to significantly improve patient outcomes compared to treatment with conventional cancer therapies such as chemotherapy, radiation, monoclonal antibodies, hormonal therapy, and photodynamic therapy.

Figure 3
Figure 3 Immune regulatory checkpoints in cancer immunotherapy. Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD1) are two well-described co-inhibitory molecules that are expressed on naïve or memory T cells and decrease antitumor immune responses. The CTLA-4-mediated immune checkpoint is induced in T cells at the initial response to antigen (early activation phase). After the T cell receptor (TCR) is triggered by antigen encounter, CTLA-4 is transported to the surface of naïve or memory T cells. In contrast, the major role of the PD1 pathway is not at the initial T cell activation stage but rather the regulation of inflammatory responses by effector T cells that recognize antigen in peripheral tissue cells. Thus, PD-1 is highly expressed by antigen-specific cytotoxic T lymphocytes (CTLs) in malignancies and is associated with impaired T-cell function. The best-characterized signal for PD1 ligand 1 (PD-L1) induction is interferon-γ (IFN-γ), which is predominantly produced by Th1 cells. Although PD-L2 expression is limited to dendritic cells (DCs) and macrophages, PD-L1 is broadly expressed in tissues and is considered a molecular shield that protects cells from auto-reactive attack. In some tumors, PDL1 is not constitutively expressed but is induced in response to inflammatory signals that are produced by an active antitumor immune response. Loading DCs with soluble PD1 decreases their function. Therefore, antibodies can be used to block inhibitory ligand:receptor interactions by acting on tumor cells, DCs (e.g., anti-PD-L1) or T cells (e.g., anti-CTLA-4 or anti-PD1). Combining the blockade of multiple inhibitory pathways synergistically decreases T cell anergy and improves T cell responsiveness against tumors.
FUTURE PERSPECTIVE

Improved treatment options that selectively target cancer-dependent pathways with little or no toxicity to normal tissues are urgently needed. Work in our laboratory focuses on these key aspects by combining the use of DCs pulsed with MHC class I and II peptides and conventional chemotherapy. Immunotherapy may be combined with conventional therapy to reduce Tregs and enhance CTL responses. Knockdown of PD-L1 and PD-L2 on monocyte-derived DCs and tumor cells may help decrease tolerance (Figure 3). DCs electroporated with PD-L small-interfering RNAs are highly effective in enhancing T cell proliferation and cytokine production and are therefore attractive candidates for improving the efficacy of DC vaccines in cancer patients[99]. Moreover, combined blockade of PD1 and CTLA-4, which play key roles in inhibiting T-cell activation, enhances antitumor immune responses compared to either agent alone (Figure 3)[100]. Combining immunotherapies, particularly agents that target different immune checkpoints, may be a promising approach. Preliminary clinical findings indicate that combined targeted therapies and simultaneous blockade of multiple immune checkpoints could promote therapeutic synergy and long-term antitumor immunity to improve clinical outcomes for melanoma patients[101]. In the metastatic CT26 CRC mouse model, simultaneous blockade of CTLA-4 and PD-L1 enhanced antitumor activity in an interleukin-15-dependent manner[102].

CONCLUSION

The limitations of surgery and adjuvant chemo/radio/antibody therapies in treating CRC patients necessitate the development of novel approaches, including immunotherapy. Despite tremendous progress in basic immunological research, effective immunotherapies for most types of cancer, including CRC, are still lacking. Immunotherapy alone may be insufficient for treating advanced CRC patients. The most promising therapeutic approach for activating antitumor immunity in cancer patients may be blockade of inhibitory immune regulatory proteins such as immune checkpoint ligands and receptors. Therefore, it is important to develop cancer vaccines that do not express inhibitory molecules such as PD-L1, but do express high levels of molecules that enhance CTL priming, such as CD80 and 4-1BBL. The blockade of multiple immune regulatory checkpoints combined with immunotherapy and/or conventional therapy may be effective in treating patients with advanced CRC.

Footnotes

P- Reviewers: Carding SR, Sakata N S- Editor: Ma YJ L- Editor: A E- Editor: Zhang DN

References
1.  Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893-2917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11128]  [Cited by in F6Publishing: 11614]  [Article Influence: 893.4]  [Reference Citation Analysis (4)]
2.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9719]  [Article Influence: 883.5]  [Reference Citation Analysis (3)]
3.  Kerr D. Clinical development of gene therapy for colorectal cancer. Nat Rev Cancer. 2003;3:615-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 68]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
4.  Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B, Starling N. Colorectal cancer. Lancet. 2010;375:1030-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1112]  [Cited by in F6Publishing: 1148]  [Article Influence: 82.0]  [Reference Citation Analysis (0)]
5.  Tebbutt NC, Cattell E, Midgley R, Cunningham D, Kerr D. Systemic treatment of colorectal cancer. Eur J Cancer. 2002;38:1000-1015.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Gallagher DJ, Kemeny N. Metastatic colorectal cancer: from improved survival to potential cure. Oncology. 2010;78:237-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 170]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
7.  Koido S, Homma S, Takahara A, Namiki Y, Tsukinaga S, Mitobe J, Odahara S, Yukawa T, Matsudaira H, Nagatsuma K. Current immunotherapeutic approaches in pancreatic cancer. Clin Dev Immunol. 2011;2011:267539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
8.  Weng D, Song B, Durfee J, Sugiyama V, Wu Z, Koido S, Calderwood SK, Gong J. Induction of cytotoxic T lymphocytes against ovarian cancer-initiating cells. Int J Cancer. 2011;129:1990-2001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
9.  Takahara A, Koido S, Ito M, Nagasaki E, Sagawa Y, Iwamoto T, Komita H, Ochi T, Fujiwara H, Yasukawa M. Gemcitabine enhances Wilms’ tumor gene WT1 expression and sensitizes human pancreatic cancer cells with WT1-specific T-cell-mediated antitumor immune response. Cancer Immunol Immunother. 2011;60:1289-1297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
10.  Weng D, Song B, Koido S, Calderwood SK, Gong J. Immunotherapy of radioresistant mammary tumors with early metastasis using molecular chaperone vaccines combined with ionizing radiation. J Immunol. 2013;191:755-763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
11.  Boon T, Coulie PG, Van den Eynde B. Tumor antigens recognized by T cells. Immunol Today. 1997;18:267-268.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Steinman RM. The dendritic cell system and its role in immunogenicity. Annu Rev Immunol. 1991;9:271-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3311]  [Cited by in F6Publishing: 3360]  [Article Influence: 101.8]  [Reference Citation Analysis (0)]
13.  Finn OJ. Cancer immunology. N Engl J Med. 2008;358:2704-2715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 667]  [Cited by in F6Publishing: 625]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
14.  Toes RE, Ossendorp F, Offringa R, Melief CJ. CD4 T cells and their role in antitumor immune responses. J Exp Med. 1999;189:753-756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 355]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
15.  Waldmann TA. Immunotherapy: past, present and future. Nat Med. 2003;9:269-277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 422]  [Cited by in F6Publishing: 377]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
16.  Tanaka Y, Morita CT, Tanaka Y, Nieves E, Brenner MB, Bloom BR. Natural and synthetic non-peptide antigens recognized by human gamma delta T cells. Nature. 1995;375:155-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 755]  [Cited by in F6Publishing: 745]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
17.  Fournié JJ, Sicard H, Poupot M, Bezombes C, Blanc A, Romagné F, Ysebaert L, Laurent G. What lessons can be learned from γδ T cell-based cancer immunotherapy trials? Cell Mol Immunol. 2013;10:35-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 130]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
18.  Kabelitz D, Wesch D, Pitters E, Zöller M. Potential of human gammadelta T lymphocytes for immunotherapy of cancer. Int J Cancer. 2004;112:727-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
19.  Corvaisier M, Moreau-Aubry A, Diez E, Bennouna J, Mosnier JF, Scotet E, Bonneville M, Jotereau F. V gamma 9V delta 2 T cell response to colon carcinoma cells. J Immunol. 2005;175:5481-5488.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Trinchieri G. Biology of natural killer cells. Adv Immunol. 1989;47:187-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2111]  [Cited by in F6Publishing: 2139]  [Article Influence: 61.1]  [Reference Citation Analysis (0)]
21.  Roder JC, Pross HF. The biology of the human natural killer cell. J Clin Immunol. 1982;2:249-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 121]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
22.  Ma Z, Liu Y, Zhou X, Yu HL, Li MQ, Tomiyama-Miyaji C, Abo T, Bai XF. Research on stress-induced apoptosis of natural killer cells and the alteration of their killing activity in mouse liver. World J Gastroenterol. 2013;19:6258-6264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
23.  Cheng M, Chen Y, Xiao W, Sun R, Tian Z. NK cell-based immunotherapy for malignant diseases. Cell Mol Immunol. 2013;10:230-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 413]  [Cited by in F6Publishing: 452]  [Article Influence: 41.1]  [Reference Citation Analysis (0)]
24.  Saito S, Harada Y, Morodomi Y, Onimaru M, Yoshida K, Kyuragi R, Matsubara H, Yonemitsu Y. Ex vivo generation of highly purified and activated natural killer cells from human peripheral blood. Hum Gene Ther Methods. 2013;24:241-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
25.  Hallett WH, Ames E, Motarjemi M, Barao I, Shanker A, Tamang DL, Sayers TJ, Hudig D, Murphy WJ. Sensitization of tumor cells to NK cell-mediated killing by proteasome inhibition. J Immunol. 2008;180:163-170.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Wennerberg E, Sarhan D, Carlsten M, Kaminskyy VO, D’Arcy P, Zhivotovsky B, Childs R, Lundqvist A. Doxorubicin sensitizes human tumor cells to NK cell- and T-cell-mediated killing by augmented TRAIL receptor signaling. Int J Cancer. 2013;133:1643-1652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
27.  Siemens DR, Hu N, Sheikhi AK, Chung E, Frederiksen LJ, Pross H, Graham CH. Hypoxia increases tumor cell shedding of MHC class I chain-related molecule: role of nitric oxide. Cancer Res. 2008;68:4746-4753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 98]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
28.  Seidel UJ, Schlegel P, Lang P. Natural killer cell mediated antibody-dependent cellular cytotoxicity in tumor immunotherapy with therapeutic antibodies. Front Immunol. 2013;4:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 167]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
29.  Inaba K, Witmer-Pack M, Inaba M, Hathcock KS, Sakuta H, Azuma M, Yagita H, Okumura K, Linsley PS, Ikehara S. The tissue distribution of the B7-2 costimulator in mice: abundant expression on dendritic cells in situ and during maturation in vitro. J Exp Med. 1994;180:1849-1860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 461]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
30.  Inaba K, Pack M, Inaba M, Sakuta H, Isdell F, Steinman RM. High levels of a major histocompatibility complex II-self peptide complex on dendritic cells from the T cell areas of lymph nodes. J Exp Med. 1997;186:665-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 213]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
31.  Théry C, Amigorena S. The cell biology of antigen presentation in dendritic cells. Curr Opin Immunol. 2001;13:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 247]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
32.  Berard F, Blanco P, Davoust J, Neidhart-Berard EM, Nouri-Shirazi M, Taquet N, Rimoldi D, Cerottini JC, Banchereau J, Palucka AK. Cross-priming of naive CD8 T cells against melanoma antigens using dendritic cells loaded with killed allogeneic melanoma cells. J Exp Med. 2000;192:1535-1544.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 219]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
33.  Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol. 2005;5:296-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 902]  [Cited by in F6Publishing: 880]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
34.  Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10867]  [Cited by in F6Publishing: 10541]  [Article Influence: 405.4]  [Reference Citation Analysis (0)]
35.  Förster R, Schubel A, Breitfeld D, Kremmer E, Renner-Müller I, Wolf E, Lipp M. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999;99:23-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1812]  [Cited by in F6Publishing: 1767]  [Article Influence: 70.7]  [Reference Citation Analysis (0)]
36.  Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev. 2010;238:247-262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in F6Publishing: 413]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
37.  Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. 2006;24:209-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 719]  [Cited by in F6Publishing: 765]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
38.  Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1257]  [Cited by in F6Publishing: 1294]  [Article Influence: 86.3]  [Reference Citation Analysis (0)]
39.  Mougiakakos D, Choudhury A, Lladser A, Kiessling R, Johansson CC. Regulatory T cells in cancer. Adv Cancer Res. 2010;107:57-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 267]  [Cited by in F6Publishing: 158]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
40.  Koido S, Homma S, Hara E, Namiki Y, Takahara A, Komita H, Nagasaki E, Ito M, Ohkusa T, Gong J. Regulation of tumor immunity by tumor/dendritic cell fusions. Clin Dev Immunol. 2010;2010:516768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
41.  Koido S, Hara E, Torii A, Homma S, Toyama Y, Kawahara H, Ogawa M, Watanabe M, Yanaga K, Fujise K. Induction of antigen-specific CD4- and CD8-mediated T-cell responses by fusions of autologous dendritic cells and metastatic colorectal cancer cells. Int J Cancer. 2005;117:587-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
42.  Koido S, Hara E, Homma S, Torii A, Toyama Y, Kawahara H, Watanabe M, Yanaga K, Fujise K, Tajiri H. Dendritic cells fused with allogeneic colorectal cancer cell line present multiple colorectal cancer-specific antigens and induce antitumor immunity against autologous tumor cells. Clin Cancer Res. 2005;11:7891-7900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 72]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
43.  Goydos JS, Elder E, Whiteside TL, Finn OJ, Lotze MT. A phase I trial of a synthetic mucin peptide vaccine. Induction of specific immune reactivity in patients with adenocarcinoma. J Surg Res. 1996;63:298-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 180]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
44.  González G, Crombet T, Catalá M, Mirabal V, Hernández JC, González Y, Marinello P, Guillén G, Lage A. A novel cancer vaccine composed of human-recombinant epidermal growth factor linked to a carrier protein: report of a pilot clinical trial. Ann Oncol. 1998;9:431-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 122]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
45.  Miyagi Y, Imai N, Sasatomi T, Yamada A, Mine T, Katagiri K, Nakagawa M, Muto A, Okouchi S, Isomoto H. Induction of cellular immune responses to tumor cells and peptides in colorectal cancer patients by vaccination with SART3 peptides. Clin Cancer Res. 2001;7:3950-3962.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Moulton HM, Yoshihara PH, Mason DH, Iversen PL, Triozzi PL. Active specific immunotherapy with a beta-human chorionic gonadotropin peptide vaccine in patients with metastatic colorectal cancer: antibody response is associated with improved survival. Clin Cancer Res. 2002;8:2044-2051.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Sugiyama H. Cancer immunotherapy targeting Wilms’ tumor gene WT1 product. Expert Rev Vaccines. 2005;4:503-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
48.  Tsuruma T, Hata F, Torigoe T, Furuhata T, Idenoue S, Kurotaki T, Yamamoto M, Yagihashi A, Ohmura T, Yamaguchi K. Phase I clinical study of anti-apoptosis protein, survivin-derived peptide vaccine therapy for patients with advanced or recurrent colorectal cancer. J Transl Med. 2004;2:19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 133]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
49.  Sadanaga N, Nagashima H, Mashino K, Tahara K, Yamaguchi H, Ohta M, Fujie T, Tanaka F, Inoue H, Takesako K. Dendritic cell vaccination with MAGE peptide is a novel therapeutic approach for gastrointestinal carcinomas. Clin Cancer Res. 2001;7:2277-2284.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Speetjens FM, Kuppen PJ, Welters MJ, Essahsah F, Voet van den Brink AM, Lantrua MG, Valentijn AR, Oostendorp J, Fathers LM, Nijman HW. Induction of p53-specific immunity by a p53 synthetic long peptide vaccine in patients treated for metastatic colorectal cancer. Clin Cancer Res. 2009;15:1086-1095.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 130]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
51.  Paulsen JE, Bjørheim J, Røe J, Eide TJ, Alexander J, Gaudernack G. Effect of vaccination with mutant KRAS peptides on rat colon carcinogenesis induced by azoxymethane. Anticancer Res. 2002;22:171-175.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Okuno K, Sugiura F, Hida JI, Tokoro T, Ishimaru E, Sukegawa Y, Ueda K. Phase I clinical trial of a novel peptide vaccine in combination with UFT/LV for metastatic colorectal cancer. Exp Ther Med. 2011;2:73-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
53.  Toubaji A, Achtar M, Provenzano M, Herrin VE, Behrens R, Hamilton M, Bernstein S, Venzon D, Gause B, Marincola F. Pilot study of mutant ras peptide-based vaccine as an adjuvant treatment in pancreatic and colorectal cancers. Cancer Immunol Immunother. 2008;57:1413-1420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 76]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
54.  Mocellin S, Pilati P, Nitti D. Peptide-based anticancer vaccines: recent advances and future perspectives. Curr Med Chem. 2009;16:4779-4796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
55.  Koido S, Hara E, Homma S, Namiki Y, Komita H, Takahara A, Nagasaki E, Ito M, Sagawa Y, Mitsunaga M. Dendritic/pancreatic carcinoma fusions for clinical use: Comparative functional analysis of healthy- versus patient-derived fusions. Clin Immunol. 2010;135:384-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
56.  Celluzzi CM, Mayordomo JI, Storkus WJ, Lotze MT, Falo LD. Peptide-pulsed dendritic cells induce antigen-specific CTL-mediated protective tumor immunity. J Exp Med. 1996;183:283-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 547]  [Cited by in F6Publishing: 569]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
57.  Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2159]  [Cited by in F6Publishing: 2010]  [Article Influence: 77.3]  [Reference Citation Analysis (0)]
58.  Koido S, Kashiwaba M, Chen D, Gendler S, Kufe D, Gong J. Induction of antitumor immunity by vaccination of dendritic cells transfected with MUC1 RNA. J Immunol. 2000;165:5713-5719.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Gong J, Chen D, Kashiwaba M, Kufe D. Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat Med. 1997;3:558-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 456]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
60.  Morse MA, Deng Y, Coleman D, Hull S, Kitrell-Fisher E, Nair S, Schlom J, Ryback ME, Lyerly HK. A Phase I study of active immunotherapy with carcinoembryonic antigen peptide (CAP-1)-pulsed, autologous human cultured dendritic cells in patients with metastatic malignancies expressing carcinoembryonic antigen. Clin Cancer Res. 1999;5:1331-1338.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Liu KJ, Wang CC, Chen LT, Cheng AL, Lin DT, Wu YC, Yu WL, Hung YM, Yang HY, Juang SH. Generation of carcinoembryonic antigen (CEA)-specific T-cell responses in HLA-A*0201 and HLA-A*2402 late-stage colorectal cancer patients after vaccination with dendritic cells loaded with CEA peptides. Clin Cancer Res. 2004;10:2645-2651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
62.  Lesterhuis WJ, de Vries IJ, Schuurhuis DH, Boullart AC, Jacobs JF, de Boer AJ, Scharenborg NM, Brouwer HM, van de Rakt MW, Figdor CG. Vaccination of colorectal cancer patients with CEA-loaded dendritic cells: antigen-specific T cell responses in DTH skin tests. Ann Oncol. 2006;17:974-980.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
63.  Nair SK, Hull S, Coleman D, Gilboa E, Lyerly HK, Morse MA. Induction of carcinoembryonic antigen (CEA)-specific cytotoxic T-lymphocyte responses in vitro using autologous dendritic cells loaded with CEA peptide or CEA RNA in patients with metastatic malignancies expressing CEA. Int J Cancer. 1999;82:121-124.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Fong L, Hou Y, Rivas A, Benike C, Yuen A, Fisher GA, Davis MM, Engleman EG. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci USA. 2001;98:8809-8814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 381]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
65.  Nair SK, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol. 1998;16:364-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 323]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
66.  Jocham D, Richter A, Hoffmann L, Iwig K, Fahlenkamp D, Zakrzewski G, Schmitt E, Dannenberg T, Lehmacher W, von Wietersheim J. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet. 2004;363:594-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 286]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
67.  Berd D, Sato T, Maguire HC, Kairys J, Mastrangelo MJ. Immunopharmacologic analysis of an autologous, hapten-modified human melanoma vaccine. J Clin Oncol. 2004;22:403-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 96]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
68.  Harris JE, Ryan L, Hoover HC, Stuart RK, Oken MM, Benson AB, Mansour E, Haller DG, Manola J, Hanna MG. Adjuvant active specific immunotherapy for stage II and III colon cancer with an autologous tumor cell vaccine: Eastern Cooperative Oncology Group Study E5283. J Clin Oncol. 2000;18:148-157.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Nemunaitis J. Vaccines in cancer: GVAX, a GM-CSF gene vaccine. Expert Rev Vaccines. 2005;4:259-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
70.  Simons JW, Jaffee EM, Weber CE, Levitsky HI, Nelson WG, Carducci MA, Lazenby AJ, Cohen LK, Finn CC, Clift SM. Bioactivity of autologous irradiated renal cell carcinoma vaccines generated by ex vivo granulocyte-macrophage colony-stimulating factor gene transfer. Cancer Res. 1997;57:1537-1546.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Simons JW, Mikhak B, Chang JF, DeMarzo AM, Carducci MA, Lim M, Weber CE, Baccala AA, Goemann MA, Clift SM. Induction of immunity to prostate cancer antigens: results of a clinical trial of vaccination with irradiated autologous prostate tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer. Cancer Res. 1999;59:5160-5168.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Salgia R, Lynch T, Skarin A, Lucca J, Lynch C, Jung K, Hodi FS, Jaklitsch M, Mentzer S, Swanson S. Vaccination with irradiated autologous tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor augments antitumor immunity in some patients with metastatic non-small-cell lung carcinoma. J Clin Oncol. 2003;21:624-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 236]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
73.  Soiffer R, Hodi FS, Haluska F, Jung K, Gillessen S, Singer S, Tanabe K, Duda R, Mentzer S, Jaklitsch M. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma. J Clin Oncol. 2003;21:3343-3350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 245]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
74.  Ockert D, Schirrmacher V, Beck N, Stoelben E, Ahlert T, Flechtenmacher J, Hagmüller E, Buchcik R, Nagel M, Saeger HD. Newcastle disease virus-infected intact autologous tumor cell vaccine for adjuvant active specific immunotherapy of resected colorectal carcinoma. Clin Cancer Res. 1996;2:21-28.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Schulze T, Kemmner W, Weitz J, Wernecke KD, Schirrmacher V, Schlag PM. Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: results of a prospective randomized trial. Cancer Immunol Immunother. 2009;58:61-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 85]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
76.  Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer J. 2011;17:359-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 121]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
77.  von Mehren M, Arlen P, Tsang KY, Rogatko A, Meropol N, Cooper HS, Davey M, McLaughlin S, Schlom J, Weiner LM. Pilot study of a dual gene recombinant avipox vaccine containing both carcinoembryonic antigen (CEA) and B7.1 transgenes in patients with recurrent CEA-expressing adenocarcinomas. Clin Cancer Res. 2000;6:2219-2228.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Marshall JL, Gulley JL, Arlen PM, Beetham PK, Tsang KY, Slack R, Hodge JW, Doren S, Grosenbach DW, Hwang J. Phase I study of sequential vaccinations with fowlpox-CEA(6D)-TRICOM alone and sequentially with vaccinia-CEA(6D)-TRICOM, with and without granulocyte-macrophage colony-stimulating factor, in patients with carcinoembryonic antigen-expressing carcinomas. J Clin Oncol. 2005;23:720-731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 204]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
79.  Hörig H, Lee DS, Conkright W, Divito J, Hasson H, LaMare M, Rivera A, Park D, Tine J, Guito K. Phase I clinical trial of a recombinant canarypoxvirus (ALVAC) vaccine expressing human carcinoembryonic antigen and the B7.1 co-stimulatory molecule. Cancer Immunol Immunother. 2000;49:504-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 135]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
80.  Kaufman HL, Lenz HJ, Marshall J, Singh D, Garett C, Cripps C, Moore M, von Mehren M, Dalfen R, Heim WJ. Combination chemotherapy and ALVAC-CEA/B7.1 vaccine in patients with metastatic colorectal cancer. Clin Cancer Res. 2008;14:4843-4849.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Rosenberg SA. Cancer vaccines based on the identification of genes encoding cancer regression antigens. Immunol Today. 1997;18:175-182.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Soda H, Koda K, Yasutomi J, Oda K, Takiguchi N, Saito N, Nakajima N. Adoptive immunotherapy for advanced cancer patients using in vitro activated cytotoxic T lymphocytes. J Surg Oncol. 1999;72:211-217.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry R, Restifo NP, Hubicki AM. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science. 2002;298:850-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2258]  [Cited by in F6Publishing: 2055]  [Article Influence: 93.4]  [Reference Citation Analysis (0)]
84.  Merhavi-Shoham E, Haga-Friedman A, Cohen CJ. Genetically modulating T-cell function to target cancer. Semin Cancer Biol. 2012;22:14-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
85.  Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, Davis JL, Morgan RA, Merino MJ, Sherry RM, Hughes MS, Kammula US, Phan GQ, Lim RM, Wank SA, Restifo NP, Robbins PF, Laurencot CM, Rosenberg SA. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011;19:620-626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 667]  [Cited by in F6Publishing: 731]  [Article Influence: 52.2]  [Reference Citation Analysis (0)]
86.  Weiner LM, Murray JC, Shuptrine CW. Antibody-based immunotherapy of cancer. Cell. 2012;148:1081-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 189]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
87.  Noguchi T, Ritter G, Nishikawa H. Antibody-based therapy in colorectal cancer. Immunotherapy. 2013;5:533-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
88.  Jiang XR, Song A, Bergelson S, Arroll T, Parekh B, May K, Chung S, Strouse R, Mire-Sluis A, Schenerman M. Advances in the assessment and control of the effector functions of therapeutic antibodies. Nat Rev Drug Discov. 2011;10:101-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 224]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
89.  Pavlidis ET, Pavlidis TE. Role of bevacizumab in colorectal cancer growth and its adverse effects: a review. World J Gastroenterol. 2013;19:5051-5060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 69]  [Cited by in F6Publishing: 69]  [Article Influence: 6.3]  [Reference Citation Analysis (4)]
90.  Jiang Z, Li C, Li F, Wang X. EGFR gene copy number as a prognostic marker in colorectal cancer patients treated with cetuximab or panitumumab: a systematic review and meta analysis. PLoS One. 2013;8:e56205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
91.  Koido S, Homma S, Takahara A, Namiki Y, Komita H, Uchiyama K, Ito M, Gong J, Ohkusa T, Tajiri H. Immunotherapy synergizes with chemotherapy targeting pancreatic cancer. Immunotherapy. 2012;4:5-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
92.  Kan S, Hazama S, Maeda K, Inoue Y, Homma S, Koido S, Okamoto M, Oka M. Suppressive effects of cyclophosphamide and gemcitabine on regulatory T-cell induction in vitro. Anticancer Res. 2012;32:5363-5369.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Lake RA, Robinson BW. Immunotherapy and chemotherapy--a practical partnership. Nat Rev Cancer. 2005;5:397-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 503]  [Cited by in F6Publishing: 468]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
94.  Nowak AK, Robinson BW, Lake RA. Synergy between chemotherapy and immunotherapy in the treatment of established murine solid tumors. Cancer Res. 2003;63:4490-4496.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455-2465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5599]  [Cited by in F6Publishing: 5908]  [Article Influence: 492.3]  [Reference Citation Analysis (0)]
96.  Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443-2454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8900]  [Cited by in F6Publishing: 9309]  [Article Influence: 775.8]  [Reference Citation Analysis (0)]
97.  Zitvogel L, Kroemer G. Targeting PD-1/PD-L1 interactions for cancer immunotherapy. Oncoimmunology. 2012;1:1223-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 274]  [Article Influence: 91.3]  [Reference Citation Analysis (0)]
98.  Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, Segal NH, Ariyan CE, Gordon RA, Reed K. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3192]  [Cited by in F6Publishing: 3187]  [Article Influence: 289.7]  [Reference Citation Analysis (0)]
99.  Hobo W, Maas F, Adisty N, de Witte T, Schaap N, van der Voort R, Dolstra H. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells. Blood. 2010;116:4501-4511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
100.  Drake CG. Combination immunotherapy approaches. Ann Oncol. 2012;23 Suppl 8:viii41-viii46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
101.  Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8693]  [Cited by in F6Publishing: 9339]  [Article Influence: 778.3]  [Reference Citation Analysis (0)]
102.  Yu P, Steel JC, Zhang M, Morris JC, Waldmann TA. Simultaneous blockade of multiple immune system inhibitory checkpoints enhances antitumor activity mediated by interleukin-15 in a murine metastatic colon carcinoma model. Clin Cancer Res. 2010;16:6019-6028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 152]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]