Open access peer-reviewed chapter

Immunotherapy for Treatment of Cancer

Written By

Aida Karachi

Submitted: 30 April 2018 Reviewed: 26 August 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.81150

From the Edited Volume

Current Trends in Cancer Management

Edited by Liliana Streba, Dan Ionut Gheonea and Michael Schenker

Chapter metrics overview

1,304 Chapter Downloads

View Full Metrics

Abstract

Cancer is known to be second cause of death worldwide despite aggressive therapeutic measures such as surgical resection of tumors, radiation therapy, and chemotherapy. The failure of currently available therapeutics for cancers, has led to increasing interest in alternative approaches including immunotherapy. Immunotherapy for cancer treatment is enhancing immune responses to fight cancer cells. Monoclonal antibodies, immune checkpoint blockades, targeted therapy, adoptive cell therapy, CAR T cells, and cancer vaccines are the most current and efficient parts of immunotherapy armamentarium. Immunotherapy has tremendous success in the treatment of cancers and is considered as a standard care of treatment or recurrence preventive therapy for variety of cancers. In this chapter, we discuss different types of immunotherapy for cancer treatment in detail.

Keywords

  • immunotherapy
  • immune checkpoint blockades
  • cancer vaccines
  • adoptive cell therapy
  • CAR T cell
  • personalized immunotherapy

1. Introduction

Cancer is the second most common cause of death in the world that has threatened health for thousands of years. Several aggressive measures such as surgical resection of tumors, chemotherapy, and radiotherapy are used to cure cancers. Although these therapeutics can minimize and inhibit cancer cells proliferation and metastasis, they have not been able to effectively defeat cancers until now. The efficacy of conventional treatments for cancer management is limited by factors such as recurrence of tumors and severe toxicities induced by therapeutics. Immunotherapy has become a tempting approach a long time after William Coley described the first immune stimulation by live bacteria for the treatment of cancer in 1893 [1]. Immunotherapy harnesses patients’ own immune system to kill cancer cells thereby reducing toxic effects of traditional chemotherapy and radiotherapy. Immune cells can identify cancer cells by recognizing tumor-associated antigens. The ability of cancer cells to escape from immune system has limited the efficacy of immunotherapy. Current novel approaches have been involved in immunotherapy to stop immune evasion of cancer cells.

Immunotherapy includes several therapies such as monoclonal antibodies, tumor cell vaccines, immune cell vaccines, and adoptive cell therapy. Monoclonal antibodies, which block cytotoxic T lymphocyte-associated protein-4 (CTLA-4), programmed cell death-1 (PD-1), dendritic cell vaccines, and chimeric antigen receptor (CAR) T cells have shown a tremendous success in clinical trials for several cancers. It is shown that immunotherapy has the potential to move to the front-line of therapeutic options in most cancers. Despite the benefits of immunotherapy, some treatments have severe side effects such as nausea, fever, and diarrhea [2]. The aim of this chapter is to study the concept of immunotherapy for cancer treatment and to provide a thorough review on immunotherapy’s developments for both oncologists and cancer immunologists.

Advertisement

2. Monoclonal antibodies

One of the mechanisms of immune system to defeat pathogens or cancers is to identify foreign substances or malignancies and generate antibodies against them. These antibodies can recognize pathogens and cancer cells by the antigens expressed on their surface. Antibodies have the ability to attach to the specific antigens and destroy foreign particles or malignancies. In the laboratory, scientists can generate many copies of antibodies that are specific to certain antigens on cancer cells. These are known as monoclonal antibodies. In 1997, the first monoclonal antibody, rituximab, was approved for treatment of non-Hodgkin’s lymphoma. Beneficial outcomes of rituximab treatment resulted in emergence and development of monoclonal antibodies as a therapeutic approach for various hematological and solid cancers [3]. The most important step in generating monoclonal antibodies for cancer treatment is identifying right antigens on cancer cells. High mutation capacity of cancer cells and existence of various antigens make this task challenging. So far, monoclonal antibodies therapy has been more beneficial against some cancers than others.

Monoclonal antibodies can defeat cancer in different ways. Some monoclonal antibodies can recognize antigens expressed by cancer cells and mark them as a target that should be destroyed by immune system. This monoclonal antibody treatment is also known as targeted therapy [4]. Some of monoclonal antibodies cause apoptosis in cancer cells by directly attaching to the cancer cells. Preventing cell proliferation, destroying cell membrane, delivering radiation or chemotherapy to cancer cells, and inhibiting blood vessel growth are other functions of monoclonal antibodies to stop cancer cells. Monoclonal antibodies can robust, mimic or maintain the immune system’s response on cancer cells in different ways, and some particular monoclonal antibodies act by more than one function [3]. Monoclonal antibodies can be categorized to three groups such as naked monoclonal antibodies (Table 1), conjugated monoclonal antibodies (Table 2), and bispecific monoclonal antibodies. Naked monoclonal antibodies act by just a single function. This single function can either be directly affecting cancer cells or by improving immune system against cancer cells. Trastuzumab is an example of monoclonal antibodies with direct effect on cancer cells. Trastuzumab can identify and block HER2 antigen, which is highly expressed on breast and stomach cancer cells. HER2 antigen is responsible for growth and proliferation of cancer cells. By blocking HER2 antigens, cancer cells are not able to expand and proliferate and spread in the body [5]. Immune check point inhibitors are monoclonal antibodies which improve immune system function. This group of antibodies will be discussed in detail later on this chapter. Some monoclonal antibodies can trigger immune system by attaching to immune cells and activating immune cells to destroy cancer cells. Alemtuzumab, which is a monoclonal antibody to treat chronic lymphocytic leukemia, binds to CD25 marker on the surface of lymphocytes and attracts immune cells to destroy cancer cells [6]. Conjugated monoclonal antibodies, also known as tagged antibodies or loaded antibodies, are antibodies that are being used to deliver either chemotherapy drugs or radioactive particles to cancer cells. These monoclonal antibodies reduce the toxic effects of systemic chemotherapy and radiotherapy by directly homing the toxic drugs to tumor microenvironment [7, 8]. Ibritumomab tiuxetan is a radio-immunotherapeutic drug which directly delivers radio isotopes to cancerous B cells in non-Hodgkin lymphoma. Ibritumomab tiuxetan is a radio-labeled monoclonal antibody against CD20 antigen, which is expressed on B cell surface. By attaching Ibritumomab tiuxetan to CD20 on the B cells and killing cancer cells, the drug is able to eliminate lymphoma [7]. Chemolabeled antibodies are monoclonal antibodies that are attached to chemotherapy drugs. Brentuximab vedotin is a chemolabeled monoclonal antibody specific for CD30 antigen on lymphocytes that delivers monomethyl auristatin E chemotherapy to cancer cells for treatment of Hodgkin lymphoma and anaplastic large cell lymphoma [9]. Ado-trastuzumab emtansine is another chemolabeled antibody attached to Mertansine (DM1) chemotherapy with ability to target HER2 molecules on breast cancer cells [10]. Immunotoxin monoclonal antibodies are a new class of monoclonal antibodies that are attached to highly toxic protein molecules of a plant or bacteria. Immunotoxins can specifically bind to their target and deliver potent toxins to cancer cells [11]. The most recent group of antibodies is bispecific monoclonal antibodies that consist of two separate antibodies targeting different specific antigens. Blinatumomab is a bispecific monoclonal antibody with the ability to bind to CD19 on lymphoma and leukemia cells and CD3 on T cells. This antibody is usually used for treatment of acute lymphocytic leukemia. By binding to two antigens on separate cells, Blinatumomab is able to bring immune cells and cancer cells together and ease the pathway for immune cells to find, attack, and kill cancer cells [12].

Monoclonal antibody Target Type Approval year Cancer
Rituximab CD20 Chimeric IgG1 1997 B cell non-Hodgkin lymphoma
Trastuzumab EGF Humanized IgG1 1998 Breast cancer
Gemtuzumab Ozogamicin CD33 2000 Acute myeloid leukemia
Alemtuzumab CD52 Humanized IgG1 2001 B cell chronic lymphocytic leukemia
Ibritumomab Tiuxetan CD20 2002 B cell non-Hodgkin lymphoma
Cetuximab VEGFR Chimeric IgG1 2004 Merkel cell carcinoma
Bevacizumab VEGF Humanized IgG1 2004 Colon cancer
Panitumumab EGFR Human IgG2 2006 Colorectal Ca
Catumaxomab CD3 Chimeric mouse-rat hybrid 2009 Malignant ascites
Ofatumumab CD20 Human IgG1 2009 B cell chronic lymphocytic leukemia
Ipilimumab CTLA-4 Human IgG1 2011 Melanoma
Brentuximab Vedotin CD30 2011 Hodgkin lymphoma
Pertuzumab HER2 Humanized IgG1 2012 Breast cancer
Ado-Trastuzumab Emtansine HER2 Humanized IgG1 2013 Breast cancer
Obinutuzumab CD20 2013 B cell chronic lymphocytic leukemia
Denosumab Human IgG2 2013 Osteoclastoma
Ramucirumab VEGFR2 Human IgG1 2014 Gastric Ca
Pembrolizumab PD-1 Humanized IgG1 2014 Melanoma
Nivolumab PD-1 Human IgG1 2014 Melanoma
Dinutuximab GD2 Chimeric IgG1 2015 Neuroblastoma
Daratumumab CD38 Human IgG1 2015 Multiple myeloma
Necitumumab EGFR Human IgG1 2015 Lung cancer
Elotuzumab SLAMF7 Humanized IgG1 2015 Multiple myeloma
Atezolizumab PD-L1 Humanized IgG1 2016 Urothelial cancer
Avelumab (14) PD-L1 human IgG1 monoclonal antibody 2017 Metastatic merkel cell carcinoma
Durvalumab PD-L1 human IgG1 kappa monoclonal antibody 2018 Urothelial carcinoma/non-small cell lung cancer

Table 1.

Unconjugated monoclonal antibodies currently approved by the Food and Drug Administration (FDA) for cancer therapy.

Monoclonal antibody Target Type Approval year Cancers
Ibritumomab tiuxetan CD20 Radionucleotide (Yttrium90 or Indium111) Murine IgG1 2002 B cell non-Hodgkin's lymphoma/lymphoproliferative disorder
Ositumomab CD20
Radionucleotide (Iodine131)
Murine IgG2a 2003 Non-Hodgkin lymphoma
Brentuximab vedotin CD30 Chimeric IgG1 Drug (auristatin E) 2011 Hodgkin lymphoma and systemic anaplastic large cell lymphoma
Trastuzumab emtansine Trastuzumab
DM1
Humanized IgG1
Drug (mertansine)
2013 Breast cancer
Tositumomab;
Iodine I 131 Tositumomab
CD19+ CD3 Murine IgG2a 2014 Acute lymphoblastic leukemia
Arcitumomab Diagnostic Murine IgG1 Colorectal cancer
Capromab pendetide Diagnostic Murine IgG1 Prostate cancer

Table 2.

Conjugated monoclonal antibodies currently approved by the Food and Drug Administration (FDA) for cancer therapy.

Based on the genetically engineering techniques, four groups of monoclonal antibodies have been developed. Murine monoclonal antibodies, which were derived from mice, were the first generation of antibodies. They were quickly eliminated from clinical studies as they were not able to interact with human immune system. Chimeric monoclonal antibodies are another category of monoclonal antibodies, consist of constant regions mostly derived from human source and variable regions entirely derived from murine source [13]. There is a subtype of chimeric non-humanized monoclonal antibodies also known as rat-mouse hybrid monoclonal antibodies with murine Fc portion that have specificities for binding to three different tumor cells, T cells and also accessory cells [14]. On the other hand, chimeric humanized monoclonal antibodies, that comprise human Fc portion, are developed with more efficient interaction with human immune system and less immunogenicity [15]. Less immunogenic and more efficient monoclonal antibodies have been developed as humanized monoclonal antibodies, which predominantly originated from human source excluding Fab portion which is derived from murine source. Human monoclonal antibodies that are fully human and are derived from transgenic mice known to be the most efficient and the least immunogenic [16].

Although monoclonal antibodies are being used for treatment of cancer, they may increase the risk of immune reactions or adverse effects. The immune reactions including acute anaphylactic reaction, serum sickness, or cytokine release syndrome (CRS) generally occur after first infusion of monoclonal antibodies. Adverse effects of monoclonal antibodies are the result of immunodeficiency mediated by blockade of specific targets. Infections such as reactivation of tuberculosis or progressive multifocal leukoencephalopathy, autoimmune diseases such as lupus and thyroid disease, cancer, dermatitis, and organ-specific adverse effects are other risks of monoclonal antibodies administration [13]. The other problem of monoclonal antibodies are constant mutation of cancer cells which results in formation of different or neoantigens that already available antibodies cannot function against them. Generation of different or neoantigens lead to absence of responsiveness to monoclonal antibodies. Developed genome sequencing techniques is promising for identifying neoantigens and producing monoclonal antibodies against this targets [3]. Monoclonal antibodies have been proven to remarkably shrink solid tumors, suppress malignancies, diminish metastasis, and increase overall survival in patients [17, 18]. Monoclonal antibodies are promising for treatment of cancers in both monotherapy and in combinatorial therapeutic approaches.

Advertisement

3. Immune checkpoint blockades

It was believed that cancer cells were completely resistant to immune system till 1800s when researchers reported regression or total elimination of some solid tumors in patients who had streptococcal skin infections or were infused with bacterial extracts [1, 19]. These studies were not continued until Sharma and Allison noticed that blocking of cytotoxic T lymphocyte-associated protein 4 (CTLA-4) enhances tumor killing capacity of T cells [20]. This hypothesis pops up that some bacterial or organisms’ extracts have the ability to block molecules on immune cells, known as checkpoints, which promote immune cells’ functionality against cancer cells. These observations led to more in-depth studies to identify immune checkpoints which their blockade can trigger robust anticancer immune responses.

One type of monoclonal antibodies that bind to immune check points is referred as immune checkpoint blockades. Checkpoints or coinhibitory receptors are molecules on immune cells that bind to their ligands expressed on normal cells. Under normal circumstances, immune checkpoints recognize healthy cells as non-pathogenic by binding to the ligands on normal cells and prevent activity of the immune system against its own tissue. Some cancer cells express check points ligands which help them to escape from recognition and elimination by immune system. By blocking immune checkpoints, immune cells gain a robust response against cancer cells. Immune check point blockades have been proven to be effective in many cancers and are promising because they are targeting immune cells by removing inhibitory pathways [21].

CTLA-4 is a coinhibitory receptor on T cells that prevent T cells activation. During T cells activation, antigen-presenting cells (APCs) present processed antigens on their major histocompatibility complex (MHC) molecules to T cell receptors. After the initial phase of activation, B7-1 or B7-2 molecules of APCs attach to CD28 on T cells. TCR signal and costimulatory B7-CD28 induce complete T cell activation that result in cytokine release from activated T cells [22]. Besides, inhibitory signals induce by CTLA-4 act in an opposite way [23]. CTLA-4 molecule expressed on T cells has a higher affinity to bind to B7 compare to CD28. In a competition between CD28 and CTLA-4, CTLA-4 predominantly binds to B7 and generates an inhibitory signal during T cells activation. Inhibitory signals of CTLA-4 halt T cells activation and induce immune tolerance. Blocking of CTLA-4 by Ipilimumab (CTLA-4 blockade) was first approved by FDA due to success of CTLA-4 blockade in treatment of melanoma patients [24]. Ipilimumab boosts immune responses to cancer cells mediated by T cells activation. Most of patients experience Ipilimumab-related side effects like diarrhea, vomiting, skin rashes, nausea, and even life-threatening effects. All patients receiving this drug are always monitored closely and side effects are managed by corticosteroids [25].

In cancer, T cells are constantly exposed to antigen stimulation which result in gradual deterioration of their function by losing cytokine production ability and persistent increase in expression of inhibitory receptors. Defects in T cell activation, cytokine production, and proliferation is defined as exhaustion. Inhibitory receptors are highly expressed on exhausted T cells. Cancer cells have a high expression of inhibitory ligands that increase the chance of exhaustion in T cells. Programmed cell death-1 (PD-1) is an inhibitory molecule known as the receptor for cell death and have regulatory inhibitory role in activation of T cells. Physiologically, PD-1/PD-1 ligand (PD-L1) signaling pathway is a way to control excessive inflammation to protect normal tissues by induction of immune tolerance [26]. Interaction of PD-1 and PD-L1, which is highly expressed on tumor cells, causes exhaustion and dysfunctionality in T cells that avoid immune response against cancer cells. PD-1 or PD-L1 inhibitors pharmacologically prevent interaction of these molecules and efficiently maintain T cells function and facilitate them to kill tumor cells. Both PD-1 and PD-L1 immune checkpoint blockades have been proven to be effective for many malignancies but still it is not obvious that whether blocking of PD-1 on T cells or PD-L1 on tumors is more effective for cancer treatment. Patients’ characteristics such as type of tumor, mutation burden of tumor, and metastases of tumor affect efficacy of PD-1/PD-L1 inhibitors [27]. PD-L1 is not constantly expressed on different tumors and even in different stages of tumor growth. Therefore, efficacy of PD-L1 blockade depends on the type of tumor, stage of tumor, location of the tumor, and many other factors [28, 29]. Atezolizumab, the first FDA-approved PD-L1 blockade, has been used as the first-line treatment of metastatic non-small lung carcinoma and cisplatin-resistant metastatic urothelial carcinoma. Avelumab, is another FDA-approved PD-L1 blockade for metastatic merkel cell carcinoma that lack efficient response to chemotherapy [30]. Nivolumab and Pembrolizumab are PD-1 blockers and are successfully used in Phase I clinical trial on patients with non-small-cell lung cancer and renal cell carcinoma. Nivolumab was approved by FDA for treatment of advanced melanoma patients after significant improved response in phase III trial. Also, Pembrolizumab is the first-line immune checkpoint blockade for the treatment of metastatic melanoma and metastatic non-small cell lung cancer [31]. These drugs have significantly increased survival of patients with minimal side effects in other solid tissue tumors. To improve benefits from immune checkpoint blockades, combinatorial strategies are under study. Combination regimens include administration of two immune checkpoint blockades together or a monoclonal antibody with chemotherapy or radiotherapy [32]. Combinatorial strategies enhance anticancer responses because each treatment works through targeting different pathways. Combination therapy of Ipilimumab/Nivolumab is approved by FDA for treatment of melanoma [33]. Pembrolizumab plus chemotherapy (pemetrexed/carboplatin) is approved for treatment of non-small cell lung carcinoma [34]. Several combination therapies including either two different checkpoint blockades or with chemo/radiotherapy are under investigation [32].

Immune checkpoint blockades have changed the treatment strategies for cancer with dramatic improves in many cancers. PD-1, PD-L1, and CTLA-4 inhibitors are able to change immune responses and it may cause adverse immune reactions. These immune reactions are usually better tolerated than chemotherapy drugs but still recognition and proactive treatments should be included in the treatment strategy for patients receiving immune checkpoint blockades [35].

Advertisement

4. Cancer vaccines

Cancer vaccines are a new generation of vaccines different to traditional prophylactic vaccines which were administered to healthy people. Cancer vaccines are administered to either prevent cancer in high-risk individuals or to treat cancer in patients with malignancies. Therapeutic cancer vaccines are able to enhance immune system to attack cancer cells. Two prophylactic vaccines were approved for cancers that are caused by virus infections. One of the prophylactic vaccines is for hepatitis B virus (HBV) infection that can cause liver cancers such as cirrhosis and hepatocellular carcinoma in those who suffer from chronic infections. Another prophylactic vaccine is against human papilloma virus (HPV) that mediates cervical, anal, vaginal, vulvar, and throat cancers as well as genital warts. Until now, preventive vaccines were only available for the cancers that are caused by infections. Therapeutic vaccines are meant to enhance immune system in order to interfere with cancer cells, stop their growth and proliferation, and kill cancer cells. Therapeutic cancers are divided to several categories of cell vaccines, peptide vaccines, and genetic vaccines.

Tumor cell vaccines are a type of cell vaccines including autologous tumor cell vaccines and allogenic tumor cell vaccines. Autologous tumor cell vaccines are isolated from patient-derived tumor cells and prepared in vitro for administration to the patient from whom the tumor cells were isolated. Preparation of tumor cells for vaccination includes irradiation of tumor cells or combining tumor cells with an immune stimulatory adjuvant such as recombinant granulocyte monocyte-colony stimulating factor (GM-CSF) [36]. Autologous cell vaccines are able to present a wide range of tumor-associated antigens to cytotoxic T cells, resulting in a robust antitumor activity. Modification of autologous tumor cells to induce higher levels of immune stimulation has been studied by many researchers. Autologous tumor cell vaccines in animal tumor models of lymphoma and melanoma were more potent when tumor cell vaccines were infected with Newcastle disease virus [37]. In another study, tumor cell vaccines were genetically modified to express higher levels of IL-2 which induced activation of T cells and natural killer (NK) cells [38]. Autologous tumor cell vaccines transduced with GM-CSF, named GVAX, are able to get involved with dendritic cells (DCs), and induce maturation of DCs. GVAX-mediated matured DCs activate cytotoxic T cells and improve T cells response to cancer [39]. Autologous tumor cell vaccines have been extensively investigated in clinical and preclinical studies on several cancers and approximately 20% of patients survived for a long time [40]. The advantage of autologous tumor cell vaccines is that the vaccines can target the patient’s own tumor-associated antigens and excludes the step to select specific antigens. One major problem in preparing autologous tumor cell vaccines is the time-consuming process of harvesting sufficient amount of tumor cells, which is a restriction for certain tumors. Appose to autologous tumor cell vaccines, allogeneic tumor cell vaccines are easy and less expensive to produce in large scales. Allogeneic whole tumor cell vaccines consist of at least two human tumor cell lines and have unlimited tumor-specific antigens. Canavaxin is an allogeneic tumor cell vaccine consisting of three irradiated allogeneic melanoma cell lines combined with adjuvant Bacillus Calmette-Guérin (BCG). Despite Canavaxin increased overall survival of melanoma patients in phase II of trials, clinical trials were terminated because of failure of the vaccine in stages III and IV [41]. Allogeneic GVAX vaccine has been studied for treatment of prostate cancer [42], breast cancer [43], and pancreatic cancer [44]. Combination of GVAX vaccine with CTLA-4 antibody (Ipilimumab) was approved by FDA for treatment of metastatic melanoma [45]. Belagenpumatucel-L is another allogeneic tumor cell vaccine formed from four non-small cell lung carcinoma (NSCLC) cell lines transfected with plasmid containing a transforming growth factor (TGF)-beta2 antisense transgene. This genetically modified vaccine secretes TGF-beta and is used for treatment of NSCLC [46].

Advertisement

5. Dendritic cell vaccines

Dendritic cell (DC) vaccines emerged as a potent cancer vaccine. DCs are professional antigen-presenting cells (APCs) that act as a bridge between innate and adoptive immune system [47]. DCs uptake pathogens, process them, and present pathogen antigens on their MHC molecules. Processed antigens on DCs are directly recognized by T cells which induce antigen-specific immune responses. Different subtypes of DCs exist in human body based on CD8, CD103, or CD11b expressions. DCs are in both non-lymphoid organs and lymphoid organs such as lymph nodes, spleens, and bone marrow. Classical DCs (cDCs) are divided to CD8+, CD103+, and CD11b+ DCs. Non-classic DCs include monocyte-derived DCs, plasmacytoid DCs, and Langerhans cells. These categories are based on expression of molecules and the location of DCs in body [48]. Studies showed that different subsets of DCs can prime and expand various T cells. For example, CD8+ CD205+ DCs present antigens on both MHC-I and MHC-II and are able to prime CD4+ T cell and CD8+ T cells but CD8-33D1+ DCs present antigens just on MHC-II and prime CD4+ T cells [49]. DCs act as a double-edged sword that can induce both immune tolerance and immune activation depending on which receptors on DCs are engaged [50]. Maturation and migration of DCs play a critical role in characteristics of DCs [51]. Matured DCs migrate to lymphoid organs and prime T cells to enhance antitumor responses. Loading of MHC molecules with cancer antigens, up regulation of costimulatory molecules such as CD40, CD80, and CD86 on DCs, and cytokine production of DCs are critically required for activation of T cells DCs [52, 53]. DC vaccines include ex vivo generation of DCs from CD34+ hematopoietic progenitor cells or peripheral blood-derived monocytes (PBMC) [53]. Ex vivo-generated DCs are loaded with appropriate source of tumor antigens and are subsequently activated with adjuvants and are administered back to patients to kill tumors. Tumor antigens derived from total tumor [54], DNA/RNA virus [55], tumor proteins, or peptides [56, 57] are utilized for DC vaccines. Moreover, some DC vaccines are composed of fusion of tumor cells and ex vivo-generated DCs [58]. Autologous DC vaccine pulsed with HLA-A0201 peptide (prostate-specific antigen) was among the first dendritic cell vaccines used in clinical trials with promising results [56]. DC vaccines have been studies in many clinical trials on various cancers. FDA-approved Sipuleucel-T DC vaccine for the first time for the treatment of metastatic castrate-resistant prostate cancer [59]. Sipuleucel-T composed of PBMC-derived DCs loaded with PA2024 (prostatic acid phosphate) fused to GM-CSF, which significantly increased patients survival. Although DC vaccines were successful in prostate cancer treatment, their efficacy in other cancers was modest. Researchers conduct studies to enhance DC vaccines potency by modulating stimulatory and inhibitory molecules on DCs. Modulation of costimulatory molecules such as CD40L, CD70, GITRL, CD137L, and OX40L [60, 61, 62, 63] or inflammatory markers of IL-12p70, IL-18, IL-12, CXCL10, and CCR7 on DCs improve DCs maturation and T cell priming characteristics [64, 65, 66, 67, 68]. The other way to enhance anticancer T cell response by DCs is to suppress inhibitory molecules on DCs. Genetically silencing of ubiquitin-editing enzyme A20 [69], suppressor of cytokine signaling 1 (SOCS1) [70], and scavenger receptor SRA/CD204 [71] improve DCs function and subsequently enhance T cell response to cancer cells.

Two of the most important limitations of cancer cell vaccines and DC vaccines are limited source of specimen and complicated procedure to generate these vaccines. New vaccines generated by tumor-associated antigen peptides combined with an adjuvant seemed to solve the restrictions of cancer cell and DC vaccines. The first encoded human tumor-associated antigen peptide was named MAGE-1 [72]. Different types of tumor-associated antigen peptides are studied. Cancer testis antigens are a group of genes available in both healthy and cancerous tissues. These genes such as MAGE, BAGE, NY-ESO-1, and SSX-2 are scant in normal tissues but are highly expressed in tumors [73, 74, 75]. Tissue differentiation antigens are available and active in both healthy tissues and tumors-like PSA and PAP in prostate cancer [76, 77], gp100, Melan-A/Mart-1, and tyrosinase in melanoma [78, 79, 80], and mammaglobin-A in breast carcinomas [81]. Tumor-specific antigens or -mutated oncogenes are a group of antigens expressed on both normal tissues and tumors with a unique up regulation in tumors such as CEA [82], MUC-1 [83], HER2/Neu [84], and certain antiapoptotic proteins (i.e. livin and survivin) [85, 86]. Clinical trials mostly focused on effects of peptide vaccines that target cancer testis antigens, and differentiation-associated antigens. To produce an effective peptide vaccine, addition of immune stimulatory adjuvant is required for an efficient immune response as tumor-associated antigens are not immunogenic. Some adjuvants used for peptide vaccine generation are aluminum salt, pathogen-associated molecular patterns (PAMPs), TLR agonists [87], BCG [88], and monophosphoryl lipid A (MPL) [89]. Cervarix is the first peptide vaccine for human papillomavirus composed of MPL and aluminum salt [90]. The advantage of peptide vaccines to DC vaccines and cancer cell vaccines is that peptide vaccines are more cost effective, but they may also appear to be less potent because they only target one or few epitopes of tumor-associated antigens. Formulation of peptide vaccines, route of delivery, and selection of immunogenic adjuvants can influence efficacy of peptide vaccines [91].

Advertisement

6. Genetic vaccines

Genetic vaccines are another approach for carrying tumor-associated antigens to patients by utilizing plasmid DNA vectors. Genetic vaccines transfect DCs and directly present tumor-associated antigens to cytotoxic T cells or they can transfect somatic cells and indirectly cross prime T cells. Each genetic vaccine can deliver many tumor-associated antigens to patients and induce a robust anticancer immunity [92]. DNA vaccines are composed of bacterial plasmids that carry genes of interest under the control of mammalian promoter. DNA vaccines are able to initiate innate immunity and based on the site of delivery, they can trigger cellular and humoral immunity [93]. Usually the transgene is cytomegalovirus (CMV) immediate early promoter and its intron A sequence [94]. Optimizing codon usage can increase the transduction of antigens. In the intra muscular administration of DNA vaccines, DNA plasmids transfect both myocytes and DCs. The plasmids act as an immunogenic and activate T cells via toll-like receptors [95]. DNA sensors in cytosol of cells such as DAI, H2B, IFI16, DDX41, LRRFIP1, and cGAS are able to detect presence of DNA vaccines. DNA sensors send signal to STING-TBK1 signaling cascade and activate interferon regulatory factor 3 which results in expression of type I interferons. TLR9 recognizes unmethylated CpG DNA and activates interferon regulatory factor 7 that induce expression of interferons. DCs phagocyte antigen-expressing cell (myocytes) and cross present antigens on MHC-I to CD8 T cells. Moreover, interferons promote this pathway. If DNA vaccines directly transfect DCs, DCs are able to uptake, process, and present antigens on MHC-I to CD8 T cells [96]. Transfection of the vector with multiple gene sequences increases the immunization and induces humoral [97] and CD8 T cell response [98]. Combination of DC vaccines with other immune stimulatory agents such as TLR agonists [99], or monoclonal antibodies [100] increase anticancer immunity. RNA vaccines are safe vaccines compared to DNA vaccines as they degrade and clear quickly in body. Total tumor RNAs are isolated from tumor tissues and they can induce a potent immune response. RNA vaccines are composed of various tumor antigens which reduce the possibility of immune escape by tumor cells. The first use of RNA vaccines was to immunize patients with mRNAs that encode tumor-associated antigens. Furthermore, RNA vaccines can be produced for personalized cancer treatment. Patients’ neoantigens can be identified by tumor exome analyzing and personalized RNA vaccine can be specifically generated. In addition to direct use of mRNAs for vaccine generation, RNAs are utilized in cell therapies. Transfecting patient-derived cells with RNAs and giving manipulated cells back to patients are another form of utilizing mRNAs. For example, transfection of patient-derived DCs with mRNA of tumor-associated antigens can induce an antigen-specific T cell response in cancer patients. Transfection of patient-derived T cells with mRNA of chimeric antigen receptors, triggers T cells to identify specific antigens on cancer cells which quickly deteriorate cancer [101]. Liposomes and protaminase are adjuvants of RNA vaccines and help to stabilize RNAs [102].

Advertisement

7. Adoptive cell therapy

Adoptive T cell therapy (ACT) is a treatment that enhances T cells’ ability to kill cancer cells by transferring immune system-derived cells to patients. The cells used for ACT can originate from the same patient or another individual. In 1988, the first ACT reduced metastatic melanoma tumors with transferring of autologous CD4+ and CD8+ tumor infiltrating lymphocytes (TILs) to the patients [103, 104]. Both peripheral blood T cells and TILs extracted from tumors are utilized to generate specific T cells for ACT. These T cells can be modified and then transferred to patients or directly administered in their natural state. TILs by their own nature have an antitumor activity as they are specific for tumor cells. TILs can recognize tumor antigens such as cancer germline antigens, neoantigens, and viral proteins and kill cancer cells [104]. After tumors are resected, the tumor tissues digest into fragments and each fragment is cultured in the presence of IL-2. The T cells are expanded and each clone is monitored for its reactivity against tumor cells. Proliferating lymphocytes kill tumor cells and produce a pure population of T cells. Cancer reactive T cells are infused back to patients. Moreover, T cells that express a TCR specific for tumor antigens can be selected in vitro from peripheral blood and expanded. Antigen-specific T cells are selected by coculturing of T cells with APCs loaded with tumor particles such as RNAs. By expansion of antigen-specific T cells, a specific antitumor T cell clone can be generated [105]. T cells with TCR targeting tumorigenic mutations such as Ras mutations have shown promise in cancer treatment. Ras is commonly mutated at the onset of tumorigenesis in the dominant population of tumor cells. Targeting Ras mutations and killing tumor cells with Ras-specific ACT may have profound effects on cancers with Ras mutations [106]. TCRs targeting KRAS G12D, a common proto-oncogene encoding GTPase, have anti-tumorigenic effects on patients with colorectal cancer [107]. Also, genetically modified antitumor T cell clones can be produced by infecting T cells with viruses that carry genetically engineered TCRs [108]. TCR-transduced T cells are generated by cloning specific TCRs into a retrovirus. Patients derived PBMCs are activated with CD3 and IL-2 and are transduced with the retrovirus encoding the antigen-specific TCR. The T cells are expanded and injected back to the individuals. Peripheral blood T cells transfected with retrovirus encoding MART-1 TCR regress tumors in melanoma [103]. Genetically engineering techniques can modify TCRs to target-specific antigens. For example, T cells with modified TCRs that target NY-ESO-1, a cancer germline antigen, were successfully used as ACT for treatment of patients with synovial cell sarcoma and melanoma [109]. One major limitation of ACTs is that they induce short-lasting responses in immune system. Administration of T cells after chemotherapy increases cancer regression due to repopulation of host T cells with antigen-specific T cells. Lymphodepletion induced by chemotherapy helps T cells from ACT to proliferate during hemostatic proliferative phase and persist for months after infusion [109]. It was also shown that high doses of IL-2 therapy contribute to expansion of the transferred cells [110, 111]. The first signal in T cell activation begins with binding of TCR to MHC molecules on APCs. Furthermore, MHC expression downregulates on APCs in cancers so that they can escape immunity [112]. In 1989, first chimeric antigen receptors (CARs) were developed to avoid interaction of T cells with MHC molecules. CAR T cells are designed to identify cancer cells and attack them without mediation of APCs. As a result, CARs act independent of any stimulatory and TCR signaling. CAR composed of a ligand-binding domain and a signaling domain. Ligand-binding domain is the extracellular part of CAR that includes B cell receptor derived single chain variable fragment. The signaling domain is made of costimulatory molecules and CD3f and 1 [112]. CD19 CAR T cells were used in clinical trial for patients with refractory B cell lymphoma and hematological malignancies. No acute graft versus host disease (GVHD) has been reported in patients except for one mild chronic ocular GVHD that was observed 2 years after CAR T cells infusion [113]. In 2017, FDA-approved Tisagenlecleucel, CD19 CAR T cell, for the treatment of acute lymphoblastic leukemia (ALL). Excellent results with these trials, increased interests in CAR T cell immunotherapy approach [114, 115]. Cytokine release syndrome (CRS) is one of the side effects of CAR T Cells. CRS is a storm of inflammatory cytokines including IL-6, IL-10, and IFN-γ that happens after the infusion of CAR T cells [2]. Patients may show symptoms such as hypotension, pulmonary edema, multi-organ failure, and even CRS-related death. Treatment of CRS includes administration of corticosteroids and IL-6 blockade. Using corticosteroids for treatment of CRS symptoms is controversial as corticosteroids dramatically decrease inflammatory cytokines and mitigate CAR T cells efficacy [116]. Another problem with CAR T cells is that they cannot penetrate into solid tumors. Studies are underway to alleviate limitations of CAR T cells and improve their efficacy for treatment of solid tumors [117].

Advertisement

8. Developing personalized immunotherapy

Many cancer patients do not benefit from immunotherapies they are receiving. Recently, many studies are focusing on identifying predictive and prognostic biomarkers in cancers as a beneficial guide for treatment decisions. This will stop administration of drugs for those patients who does not benefit from them and improve treatment in patients that are most likely respond to specific immunotherapies. Selecting the appropriate immunotherapy for each cancer patient is still a challenge. Scientists and oncologists are developing methods in genomic testing to discover cell signaling and biomarkers involved in responding to immunotherapy. It has been shown that cancers identified by specific quantity or pattern of mutations in the tumor microenvironment or surrounding area are more responsive to immune checkpoint blockades. Of note, scientists are trying to exploit other drugs to alter the tumor microenvironment of less immune responsive tumors, known as cold tumors, and turn them to check point blockades susceptible tumors that are defined as hot tumors [32]. Altering tumor microenvironment and surrounding tissues can increase the number of patients who can benefit from immune checkpoint blockades. Immunopharmacogenomics approach is providing a significant hope for personalized immunotherapy [118].

Advertisement

9. Conclusion

In summary, immunotherapy shows a tremendous potential in treatment of cancer. Different immunotherapies have been approved by FDA for prevention and treatment of cancers. Despite the breakthroughs achieved by immunotherapy, many cancers still do not respond to immunotherapy. Monotherapy of immune checkpoint blockades or other immunotherapies failed in treatment of some cancers. Finding the efficient treatment by combinatorial immunotherapies or combination of immunotherapy and traditional chemotherapy and radiotherapy are under investigation. Development of DCs and cancer vaccines, immune checkpoint blockades, CAR T cells, and ACT requires an in-depth understanding of tumor microenvironment and identifying tumor-specific antigens. More studies to develop immunotherapy can provide improved efficacy in cancer treatments.

References

  1. 1. Coley WB. II. Contribution to the knowledge of sarcoma. Annals of Surgery. 1891;14(3):199-220
  2. 2. Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer Journal. 2014;20(2):119-122
  3. 3. Coulson A, Levy A, Gossell-Williams M. Monoclonal antibodies in cancer therapy: Mechanisms, successes and limitations. The West Indian Medical Journal. 2014;63(6):650-654
  4. 4. Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nature Reviews. Cancer. 2012;12(4):237-251
  5. 5. Pinto AC, Ades F, de Azambuja E, Piccart-Gebhart M. Trastuzumab for patients with HER2 positive breast cancer: Delivery, duration and combination therapies. Breast. 2013;22(Suppl 2):S152-S155
  6. 6. Fiegl M, Stauder R, Steurer M, et al. Alemtuzumab in chronic lymphocytic leukemia: Final results of a large observational multicenter study in mostly pretreated patients. Annals of Hematology. 2014;93(2):267-277
  7. 7. Steiner M, Neri D. Antibody-radionuclide conjugates for cancer therapy: Historical considerations and new trends. Clinical Cancer Research. 2011;17(20):6406-6416
  8. 8. Flygare JA, Pillow TH, Aristoff P. Antibody-drug conjugates for the treatment of cancer. Chemical Biology & Drug Design. 2013;81(1):113-121
  9. 9. Fanale MA, Horwitz SM, Forero-Torres A, et al. Brentuximab vedotin in the front-line treatment of patients with CD30+ peripheral T-cell lymphomas: Results of a phase I study. Journal of Clinical Oncology. 2014;32(28):3137-3143
  10. 10. Amiri-Kordestani L, Blumenthal GM, Xu QC, et al. FDA approval: Ado-trastuzumab emtansine for the treatment of patients with HER2-positive metastatic breast cancer. Clinical Cancer Research. 2014;20(17):4436-4441
  11. 11. Ehrlich D, Wang B, Lu W, Dowling P, Yuan R. Intratumoral anti-HuD immunotoxin therapy for small cell lung cancer and neuroblastoma. Journal of Hematology & Oncology. 2014;7:91
  12. 12. Kaplan JB, Grischenko M, Giles FJ. Blinatumomab for the treatment of acute lymphoblastic leukemia. Investigational New Drugs. 2015;33(6):1271-1279
  13. 13. Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ. The safety and side effects of monoclonal antibodies. Nature Reviews. Drug Discovery. 2010;9(4):325-338
  14. 14. Seimetz D. Novel monoclonal antibodies for cancer treatment: The trifunctional antibody catumaxomab (removab). Journal of Cancer. 2011;2:309-316
  15. 15. Reichert JM, Rosensweig CJ, Faden LB, Dewitz MC. Monoclonal antibody successes in the clinic. Nature Biotechnology. 2005;23(9):1073-1078
  16. 16. Lonberg N. Human monoclonal antibodies from transgenic mice. Handbook of Experimental Pharmacology. 2008;181:69-97
  17. 17. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. Journal of Clinical Oncology. 2014;32(10):1020-1030
  18. 18. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Annals of Oncology. 2016;27(7):1362
  19. 19. Littman DR. Releasing the brakes on cancer immunotherapy. Cell. 2015;162(6):1186-1190
  20. 20. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell. 2015;161(2):205-214
  21. 21. Tsai HF, Hsu PN. Cancer immunotherapy by targeting immune checkpoints: Mechanism of T cell dysfunction in cancer immunity and new therapeutic targets. Journal of Biomedical Science. 2017;24(1):35
  22. 22. Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunological Reviews. 2008;224:166-182
  23. 23. Collins AV, Brodie DW, Gilbert RJ, et al. The interaction properties of costimulatory molecules revisited. Immunity. 2002;17(2):201-210
  24. 24. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England Journal of Medicine. 2010;363(8):711-723
  25. 25. Camacho LH. CTLA-4 blockade with ipilimumab: Biology, safety, efficacy, and future considerations. Cancer Medicine. 2015;4(5):661-672
  26. 26. McDermott J, Jimeno A. Pembrolizumab: PD-1 inhibition as a therapeutic strategy in cancer. Drugs Today (Barc). 2015;51(1):7-20
  27. 27. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends in Molecular Medicine. 2015;21(1):24-33
  28. 28. Mu L, Long Y, Yang C, et al. The IDH1 mutation-induced oncometabolite, 2-hydroxyglutarate, may affect DNA methylation and expression of PD-L1 in gliomas. Frontiers in Molecular Neuroscience. 2018;11:82
  29. 29. Karachi A, Azari H, Flores C, Yang C, Dastmalchi F, Mitchell D, et al. TMZ results in priming of host immunity and changes in GBM tumor PDL-1 expression in a dose dependent fashion that can be leveraged for combination with immune checkpoint blockade: December 2016. Neuro-Oncology;18(Suppl 6):vi202
  30. 30. Gaiser MR, Bongiorno M, Brownell I. PD-L1 inhibition with avelumab for metastatic Merkel cell carcinoma. Expert Review of Clinical Pharmacology. 2018;11(4):345-359
  31. 31. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. Journal of Clinical Oncology. 2015;33(17):1974-1982
  32. 32. Karachi A, Dastmalchi F, Mitchell D, Rahman M. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro-Oncology. 2018; noy072. https://doi.org/10.1093/neuonc/noy072
  33. 33. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. The New England Journal of Medicine. 2015;372(21):2006-2017
  34. 34. Langer CJ, Gadgeel SM, Borghaei H, et al. Carboplatin and pemetrexed with or without pembrolizumab for advanced, non-squamous non-small-cell lung cancer: a randomised, phase 2 cohort of the open-label KEYNOTE-021 study. Lancet Oncology. 2016;17(11):1497-1508
  35. 35. Davies M, Duffield EA. Safety of checkpoint inhibitors for cancer treatment: Strategies for patient monitoring and management of immune-mediated adverse events. ImmunoTargets and Therapy. 2017;6:51-71
  36. 36. Chiang CL, Coukos G, Kandalaft LE. Whole tumor antigen vaccines: Where are we? Vaccines (Basel). 2015;3(2):344-372
  37. 37. Heicappell R, Schirrmacher V, von Hoegen P, Ahlert T, Appelhans B. Prevention of metastatic spread by postoperative immunotherapy with virally modified autologous tumor cells. I. Parameters for optimal therapeutic effects. International Journal of Cancer. 1986;37(4):569-577
  38. 38. Asada H, Kishida T, Hirai H, et al. Significant antitumor effects obtained by autologous tumor cell vaccine engineered to secrete interleukin (IL)-12 and IL-18 by means of the EBV/lipoplex. Molecular Therapy. 2002;5(5 Pt 1):609-616
  39. 39. Mach N, Gillessen S, Wilson SB, Sheehan C, Mihm M, Dranoff G. Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or Flt3-ligand. Cancer Research. 2000;60(12):3239-3246
  40. 40. Dranoff G. Targets of protective tumor immunity. Annals of the New York Academy of Sciences. 2009;1174:74-80
  41. 41. Sondak VK, Sabel MS, Mule JJ. Allogeneic and autologous melanoma vaccines: Where have we been and where are we going? Clinical Cancer Research. 2006;12(7 Pt 2):2337s-2341s
  42. 42. Small EJ, Sacks N, Nemunaitis J, et al. Granulocyte macrophage colony-stimulating factor--secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clinical Cancer Research. 2007;13(13):3883-3891
  43. 43. Emens LA, Asquith JM, Leatherman JM, 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. Journal of Clinical Oncology. 2009;27(35):5911-5918
  44. 44. Lutz E, Yeo CJ, Lillemoe KD, et al. A lethally irradiated allogeneic granulocyte-macrophage colony stimulating factor-secreting tumor vaccine for pancreatic adenocarcinoma. A phase II trial of safety, efficacy, and immune activation. Annals of Surgery. 2011;253(2):328-335
  45. 45. van den Eertwegh AJ, Versluis J, van den Berg HP, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: A phase 1 dose-escalation trial. The Lancet Oncology. 2012;13(5):509-517
  46. 46. Nemunaitis J, Nemunaitis M, Senzer N, et al. Phase II trial of Belagenpumatucel-L, a TGF-beta2 antisense gene modified allogeneic tumor vaccine in advanced non small cell lung cancer (NSCLC) patients. Cancer Gene Therapy. 2009;16(8):620-624
  47. 47. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392(6673):245-252
  48. 48. Mildner A, Jung S. Development and function of dendritic cell subsets. Immunity. 2014;40(5):642-656
  49. 49. Dudziak D, Kamphorst AO, Heidkamp GF, et al. Differential antigen processing by dendritic cell subsets in vivo. Science. 2007;315(5808):107-111
  50. 50. Karachi A, Fazeli M, Karimi MH, et al. Evaluation of immunomodulatory effects of mesenchymal stem cells soluble factors on miR-155 and miR-23b expression in mice dendritic cells. Immunological Investigations. 2015;44(5):427-437
  51. 51. Dastmalchi F, Karachi A, Azari H, Mitchell D, Rahman M. Strategy to enhance DC migration for increased efficacy of dendritic cells vaccine immunotherapy. Neuro-Oncology. 2017;19(suppl_6):vi125
  52. 52. Frankenberger B, Schendel DJ. Third generation dendritic cell vaccines for tumor immunotherapy. European Journal of Cell Biology. 2012;91(1):53-58
  53. 53. Dastmalchi F, Karachi A, Mitchell D, Rahman M. Dendritic Cell Therapy. In: eLS. Chichester: John Wiley & Sons, Ltd; 2018. DOI: 10.1002/9780470015902.a0024243
  54. 54. Batich KA, Reap EA, Archer GE, et al. Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clinical Cancer Research. 2017;23(8):1898-1909
  55. 55. Nair SK, Morse M, Boczkowski D, et al. Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor RNA-transfected dendritic cells. Annals of Surgery. 2002;235(4):540-549
  56. 56. Murphy G, Tjoa B, Ragde H, Kenny G, Boynton A. Phase I clinical trial: T-cell therapy for prostate cancer using autologous dendritic cells pulsed with HLA-A0201-specific peptides from prostate-specific membrane antigen. The Prostate. 1996;29(6):371-380
  57. 57. Schuler-Thurner B, Schultz ES, Berger TG, et al. Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. The Journal of Experimental Medicine. 2002;195(10):1279-1288
  58. 58. Rosenblatt J, Vasir B, Uhl L, et al. Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood. 2011;117(2):393-402
  59. 59. Longo DL. New therapies for castration-resistant prostate cancer. The New England Journal of Medicine. 2010;363(5):479-481
  60. 60. Bonehill A, Tuyaerts S, Van Nuffel AM, et al. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Molecular Therapy. 2008;16(6):1170-1180
  61. 61. Dannull J, Nair S, Su Z, et al. Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA encoding OX40 ligand. Blood. 2005;105(8):3206-3213
  62. 62. Grunebach F, Kayser K, Weck MM, Muller MR, Appel S, Brossart P. Cotransfection of dendritic cells with RNA coding for HER-2/neu and 4-1BBL increases the induction of tumor antigen specific cytotoxic T lymphocytes. Cancer Gene Therapy. 2005;12(9):749-756
  63. 63. Tuyaerts S, Aerts JL, Corthals J, et al. Current approaches in dendritic cell generation and future implications for cancer immunotherapy. Cancer Immunology, Immunotherapy. 2007;56(10):1513-1537
  64. 64. Iinuma H, Okinaga K, Fukushima R, et al. Superior protective and therapeutic effects of IL-12 and IL-18 gene-transduced dendritic neuroblastoma fusion cells on liver metastasis of murine neuroblastoma. Journal of Immunology. 2006;176(6):3461-3469
  65. 65. Kang TH, Bae HC, Kim SH, et al. Modification of dendritic cells with interferon-gamma-inducible protein-10 gene to enhance vaccine potency. The Journal of Gene Medicine. 2009;11(10):889-898
  66. 66. Minkis K, Kavanagh DG, Alter G, et al. Type 2 bias of T cells expanded from the blood of melanoma patients switched to type 1 by IL-12p70 mRNA-transfected dendritic cells. Cancer Research. 2008;68(22):9441-9450
  67. 67. Ogawa F, Iinuma H, Okinaga K. Dendritic cell vaccine therapy by immunization with fusion cells of interleukin-2 gene-transduced, spleen-derived dendritic cells and tumour cells. Scandinavian Journal of Immunology. 2004;59(5):432-439
  68. 68. Okada N, Mori N, Koretomo R, et al. Augmentation of the migratory ability of DC-based vaccine into regional lymph nodes by efficient CCR7 gene transduction. Gene Therapy. 2005;12(2):129-139
  69. 69. Song XT, Evel-Kabler K, Shen L, Rollins L, Huang XF, Chen SY. A20 is an antigen presentation attenuator, and its inhibition overcomes regulatory T cell-mediated suppression. Nature Medicine. 2008;14(3):258-265
  70. 70. Palmer DC, Restifo NP. Suppressors of cytokine signaling (SOCS) in T cell differentiation, maturation, and function. Trends in Immunology. 2009;30(12):592-602
  71. 71. Yu X, Yi H, Guo C, et al. Pattern recognition scavenger receptor CD204 attenuates Toll-like receptor 4-induced NF-kappaB activation by directly inhibiting ubiquitination of tumor necrosis factor (TNF) receptor-associated factor 6. The Journal of Biological Chemistry. 2011;286(21):18795-18806
  72. 72. van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643-1647
  73. 73. De Smet C, Lurquin C, van der Bruggen P, De Plaen E, Brasseur F, Boon T. Sequence and expression pattern of the human MAGE2 gene. Immunogenetics. 1994;39(2):121-129
  74. 74. Gnjatic S, Ritter E, Buchler MW, et al. Seromic profiling of ovarian and pancreatic cancer. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(11):5088-5093
  75. 75. Karbach J, Neumann A, Atmaca A, et al. Efficient in vivo priming by vaccination with recombinant NY-ESO-1 protein and CpG in antigen naive prostate cancer patients. Clinical Cancer Research. 2011;17(4):861-870
  76. 76. Correale P, Walmsley K, Nieroda C, et al. In vitro generation of human cytotoxic T lymphocytes specific for peptides derived from prostate-specific antigen. Journal of the National Cancer Institute. 1997;89(4):293-300
  77. 77. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. The New England Journal of Medicine. 2010;363(5):411-422
  78. 78. Bakker AB, Schreurs MW, de Boer AJ, et al. Melanocyte lineage-specific antigen gp100 is recognized by melanoma-derived tumor-infiltrating lymphocytes. The Journal of Experimental Medicine. 1994;179(3):1005-1009
  79. 79. Kawakami Y, Eliyahu S, Sakaguchi K, et al. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes. The Journal of Experimental Medicine. 1994;180(1):347-352
  80. 80. Parkhurst MR, Fitzgerald EB, Southwood S, Sette A, Rosenberg SA, Kawakami Y. Identification of a shared HLA-A*0201-restricted T-cell epitope from the melanoma antigen tyrosinase-related protein 2 (TRP2). Cancer Research. 1998;58(21):4895-4901
  81. 81. Jaramillo A, Majumder K, Manna PP, et al. Identification of HLA-A3-restricted CD8+ T cell epitopes derived from mammaglobin-A, a tumor-associated antigen of human breast cancer. International Journal of Cancer. 2002;102(5):499-506
  82. 82. Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton JM, Schlom J. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine. Journal of the National Cancer Institute. 1995;87(13):982-990
  83. 83. Finn OJ, Gantt KR, Lepisto AJ, Pejawar-Gaddy S, Xue J, Beatty PL. Importance of MUC1 and spontaneous mouse tumor models for understanding the immunobiology of human adenocarcinomas. Immunologic Research. 2011;50(2-3):261-268
  84. 84. Disis ML, Wallace DR, Gooley TA, et al. Concurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancer. Journal of Clinical Oncology. 2009;27(28):4685-4692
  85. 85. Schmollinger JC, Vonderheide RH, Hoar KM, et al. Melanoma inhibitor of apoptosis protein (ML-IAP) is a target for immune-mediated tumor destruction. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(6):3398-3403
  86. 86. Schmidt SM, Schag K, Muller MR, et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood. 2003;102(2):571-576
  87. 87. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34(5):637-650
  88. 88. Heldwein KA, Liang MD, Andresen TK, et al. TLR2 and TLR4 serve distinct roles in the host immune response against Mycobacterium bovis BCG. Journal of Leukocyte Biology. 2003;74(2):277-286
  89. 89. Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science. 2007;316(5831):1628-1632
  90. 90. Clarke MA, Wentzensen N, Mirabello L, et al. Human papillomavirus DNA methylation as a potential biomarker for cervical cancer. Cancer Epidemiology, Biomarkers & Prevention. 2012;21(12):2125-2137
  91. 91. Kumai T, Kobayashi H, Harabuchi Y, Celis E. Peptide vaccines in cancer-old concept revisited. Current Opinion in Immunology. 2017;45:1-7
  92. 92. Aurisicchio L, Ciliberto G. Genetic cancer vaccines: Current status and perspectives. Expert Opinion on Biological Therapy. 2012;12(8):1043-1058
  93. 93. Liu MA. DNA vaccines: An historical perspective and view to the future. Immunological Reviews. 2011;239(1):62-84
  94. 94. Stratford R, Douce G, Zhang-Barber L, Fairweather N, Eskola J, Dougan G. Influence of codon usage on the immunogenicity of a DNA vaccine against tetanus. Vaccine. 2000;19(7-8):810-815
  95. 95. Barber GN. Cytoplasmic DNA innate immune pathways. Immunological Reviews. 2011;243(1):99-108
  96. 96. Yang B, Jeang J, Yang A, Wu TC, Hung CF. DNA vaccine for cancer immunotherapy. Human Vaccines & Immunotherapeutics. 2014;10(11):3153-3164
  97. 97. Walter P, Johnson AE. Signal sequence recognition and protein targeting to the endoplasmic reticulum membrane. Annual Review of Cell Biology. 1994;10:87-119
  98. 98. Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: Precision tools for activating effective immunity against cancer. Nature Reviews. Cancer. 2008;8(2):108-120
  99. 99. Dharmapuri S, Aurisicchio L, Neuner P, Verdirame M, Ciliberto G, La Monica N. An oral TLR7 agonist is a potent adjuvant of DNA vaccination in transgenic mouse tumor models. Cancer Gene Therapy. 2009;16(5):462-472
  100. 100. Orlandi F, Guevara-Patino JA, Merghoub T, Wolchok JD, Houghton AN, Gregor PD. Combination of epitope-optimized DNA vaccination and passive infusion of monoclonal antibody against HER2/neu leads to breast tumor regression in mice. Vaccine. 2011;29(20):3646-3654
  101. 101. Fiedler K, Lazzaro S, Lutz J, Rauch S, Heidenreich R. mRNA cancer vaccines. Recent Results in Cancer Research. 2016;209:61-85
  102. 102. Fotin-Mleczek M, Zanzinger K, Heidenreich R, et al. Highly potent mRNA based cancer vaccines represent an attractive platform for combination therapies supporting an improved therapeutic effect. The Journal of Gene Medicine. 2012;14(6):428-439
  103. 103. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 2015;348(6230):62-68
  104. 104. Verdegaal EM. Adoptive cell therapy: A highly successful individualized therapy for melanoma with great potential for other malignancies. Current Opinion in Immunology. 2016;39:90-95
  105. 105. Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(25):16168-16173
  106. 106. Bryant KL, Mancias JD, Kimmelman AC, Der CJ. KRAS: Feeding pancreatic cancer proliferation. Trends in Biochemical Sciences. 2014;39(2):91-100
  107. 107. Tran E, Robbins PF, Lu YC, et al. T-cell transfer therapy targeting mutant KRAS in cancer. The New England Journal of Medicine. 2016;375(23):2255-2262
  108. 108. Morgan RA, Dudley ME, Wunderlich JR, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314(5796):126-129
  109. 109. Robbins PF, Morgan RA, Feldman SA, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. Journal of Clinical Oncology. 2011;29(7):917-924
  110. 110. Donohue JH, Rosenstein M, Chang AE, Lotze MT, Robb RJ, Rosenberg SA. The systemic administration of purified interleukin 2 enhances the ability of sensitized murine lymphocytes to cure a disseminated syngeneic lymphoma. Journal of Immunology. 1984;132(4):2123-2128
  111. 111. Dastmalchi F, Karachi A, Allison JR, Basso K, Mitchell D, Rahman M. In vivo cellular tracking with 13C labeling of adopetive transferred T cells for the treatment of brain tumors. Neuro-Oncology. 2017;19(suppl_6):vi119
  112. 112. Leone P, Shin EC, Perosa F, Vacca A, Dammacco F, Racanelli V. MHC class I antigen processing and presenting machinery: Organization, function, and defects in tumor cells. Journal of the National Cancer Institute. 2013;105(16):1172-1187
  113. 113. Brudno JN, Somerville RP, Shi V, et al. Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that Progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. Journal of Clinical Oncology. 2016;34(10):1112-1121
  114. 114. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. Journal of Clinical Oncology. 2015;33(6):540-549
  115. 115. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England Journal of Medicine. 2014;371(16):1507-1517
  116. 116. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid Leukemia; chimeric antigen receptor-modified T cells for acute lymphoid Leukemia; chimeric antigen receptor T cells for sustained remissions in Leukemia. The New England Journal of Medicine. 2016;374(10):998
  117. 117. Ninomiya S, Narala N, Huye L, et al. Tumor indoleamine 2,3-dioxygenase (IDO) inhibits CD19-CAR T cells and is downregulated by lymphodepleting drugs. Blood. 2015;125(25):3905-3916
  118. 118. Kakimi K, Karasaki T, Matsushita H, Sugie T. Advances in personalized cancer immunotherapy. Breast Cancer. 2017;24:16-24

Written By

Aida Karachi

Submitted: 30 April 2018 Reviewed: 26 August 2018 Published: 05 November 2018