Cancer: Immunology and Immunotharapy
Saloni S. Chhajed, Mayuri V. Mali, Azam Z. Shaikh, S. P. Pawar, Ritik. S. Jain
Department of Pharmaceutics P.S.G.V.P.M’s College of Pharmacy, Shahada.
*Corresponding Author E-mail: ritikbadera390@gmail.co
ABSTRACT:
During immune surveillance, the host provides defense versus foreign antigens. By targeting surface antigens expressed on tumor cells, monoclonal antibodies have demonstrated efficacy as cancer therapeutics. Recent successful antibody-based strategies have focused on enhancing antitumor immune responses by targeting immune cells, irrespective of tumor antigens. The use of antibodies to woodcut pathways inhibiting the endogenous immune response to cancer, known as checkpoint tampon therapy, has stirred up a unconfined deal of excitement among scientists, physicians, and patients alike. Clinical trials evaluating the safety and efficacy of antibodies that woodcut the T lamina inhibitory molecules cytotoxic T-lymphocyte various kinds of immunotherapy treatment for cancer are either misogynist to the public or are in the process of clinical trials. Immunotherapy treatments have the potential to treat cancer with significantly less toxicity than chemotherapy and radiation treatments. An accent on cellular infusion as a method of either enhancing the immune system by creating an environment for se-questering the host immune system to wade cancer cells or increasingly directly inserting cells to di-rectly wade cancer cells will be provided in this review. Various forms of cancer vaccines are moreover discussed in this paper as an important speciality in immunotherapy. This review seeks to describe various methodologies associated with overseeing immunotherapy in the treatment of cancer.
KEYWORDS: Cancer, Checkpoint tampon therapy, T-lymphocyte.
INTRODUCTION:
The past few years have seen a upsurge of research on the immune system yielding a increasingly understanding of how cancer progresses and offering new ways to pause it.1 In 1891. William Coley introduce cancer patients with yes-man to erupt an immune response, tideway now experiencing a renewal now immunologists are finding ways regulation the immune system, including training immune cells in realize a patient's particular cancer2,3 discovery that tumors can urgently suppress immunity has led to the minutiae of checkpoint blockades that prevent this suppression.4
Last year's Lasker DeBakey Clinical Research Award was awarded to James Allison for discovering that pathogen staying of the T lamina molecule cytotoxic T-lymphocyte associated protein 4 [CTLA-4] unleashes the body's immune response versus malignant tumors. This lead to minutiae of multiple immune checkpoint therapies that are prolonging and saving the lives of many cancer patients.5
The successful treatment of multiple mouse tumors with anti-CTLA-4 impressed many immunologists, but the perceived lack of success many older immune-based therapies produce a very upper harm for up-and-coming immune checkpoint therapy to the clinic. The turning point came with 3 trial comparing Ipilimumab to a melanoma peptide vaccine in metastatic melanoma patients who had the human leukocyte antigens [HLA] A0201 allele6 Key to the success of the study was the visualization to evaluate overall survival rather than response rate, and in the large study. Ipilimumab monotherapy resulted in increasingly than 20% long-term survival. The success of this trial led the Food and Drug Administration [FDA] to finally legitimatize Ipilimumab for the treatment of metastatic melanoma in 2011, At virtually the time when Ipilimumab was stuff considered for FDA approval, renewed excitement well-nigh immune checkpoint therapy came from clinical studies targeting a second immune inhibitory molecule. programmed lamina death 1 [PD-1] PD-1 was discovered by Tasuku Honjo in 1992 in a screen for genes expressed during programmed lamina death of a T lamina hybridoma,7 incubation of tumor cells resulting from the progressive unifying of multiple genetic.8 Alterations in tumor stroma microenvironments may moreover promotes the minutiae of tumor lamina heterogeneity through extrinsic vivification of unrepeatable tumor lamina signaling pathways.9 Moreover, recent studies have suggested that heterogeneity is a result of the hierarchical organization of tumor cells by a subset of cells with stem or progenitor lamina features known as cancer stem cells [CSC].10
Cancer Immunology:
Although originally considered as monoclonal, tumor cells show heterogeneous morphology and behavine.11,12 This heterogeneity has traditionally been explained by the clonal incubation of tumor cells resulting from the progressive unifying of multiple genetic Alterations in tumor stroma microenvironments may moreover promotes the minutiae of tumor lamina heterogeneity. through extrinsic vivification of unrepealable tumor lamina signaling pathways.13 The concept of cancer as an unwonted stem lamina disease was proposed based on the similar skills of cancer cells and normal stem cells to self-renew, produce heterogeneous. progeny and moreover divide in an unlimited maleate.14,15 However, CSC proposition has only recently been experiment validated by the identification of a subset of unrepealable self reewing stem lamina marker-positive cells with a hierarch is confir ization.16,17 Therefore, CSC are a pivotal target for the eradication of many cancers including liver cancer.18 As tumors are heterogeneous and show distinctive genetic and epigenetic profiles, there may not be a single biomarker that will prove sufficient information for predicting treatment response and patient outcome. Examples of informative tumor biomarkers are molecular features of neoplastic cells, including epidermal growth factor receptor [EGFR] mutations in lung cancer19,20; microsatellite instability [MSI] in colorectal cancer21-23; estrogen receptor 1 [ESR1], progesterone receptor [PCR] and erb-b2 receptor tymsine kinase 2 [ERBB2/HER2] expression in breast cancer24,25; transmembrane protease, serine 2 and ETS-related gene fusion [TMPRSS2-ERG] translocation. in prostate cancer26 and CpG island methylation, and kirsten rat sarcoma viral oncogene [KRAS], B-raf proto oncogene [BRAF], phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit start [PIK3CA] and tumor protein[TP]-53 mutations in multiple cancer types27-29.
Cancer immuno therapy:
For increasingly than half a century, scientists have been trying to turn the body's immune system versus cancer. But decades of failures have revealed that tumors have the worthiness to evade, tamp lanugo and overwhelm the normal immune response. Most modern immune therapies try to get the immune system to recognize and wade tumor cells.30 The wardship of monoclonal antibodies [mAb] versus tumor antigens in HER2-positive breast cancer [Trastuzumab]31. throne and neck, lung, and colorectal cancers that express EGFR32-34 Antibodies can moreover target immune cells at the tumor site to aid the vivification of effector cells and promote increasingly constructive antitumor immunity. 35 One such treatment, the vaccine sipuleucel-T [marketed as Provenge by Dendreon Corporation in Seattle], was tried by the US FDA in 2010 for use in prostate cancer, which is a move that generated a lot of excitement. But the drug has proven disappointing, with benefits limited to a small percentage of patients, Dendreon is now reported to be for sale. The problem, researchers have slowly been realizing, is that stepping on the immune system's gas pedal isn't enough. It is moreover necessary to release its brakes, and that is where immune checkpoint blockades come in. In 2011, the US FDA tried the anti-CTLA4 drug Ipilimumab [developed by BMS and marketed as Yervoy], which was based on Allison's research and sooner saved the lives of some of Ribas's patients. CTLA-4 is not the only checkpoint stuff targeted by researchers and drug developers. Early trials suggest that drugs that woodcut a variegated checkpoint, which is PD-1, are plane increasingly constructive and have fewer side effects than Ipilimumab.36 Historically, stereotype survival was six to nine months.37 Early research suggests that Ipilimumab may be plane increasingly constructive when combined with other drugs. In remoter vestige for the value of drug combinations, Ipilimumab and Nivolumab towards to complement each other. There are three strategies for ACT therapies, the most ripened of which is the simplest. The tissue surrounding a tumor is likely to contain immune cells with antitumor activity, so doctors take a sample of this tissue and select those T cells that have been primed to wade the cancer. They culture these cells in the lab until they have enough, and re infuse the cells when to patients withal with the T-cell growth factor IL-2, which promotes the proliferation of antigen specific T cells.
mAb Targetting Cancer-associated Proteins:
Specific recognition by mAb has permitted the identification of cytokines and cell-surface molecules involved in humoral antibody-mediated and cellular immune responses. Antibodies may target tumor cells by engaging surface antigen differentially expressed in cancers. For example. Rituximab target CD20 in non-Hodgkin B lamina lymphoma, Trastuzumab targets HER2 in breast cancer, and Cetuximab targets EGFR in colorectal cancer. Blocking the ligand receptor growth can evoke the tumor lamina death and survival pathways. Innate immune effector mechanisms engaging the Fc portion of antibodies via Fc receptors including complement-mediated cytotoxicity [CMC] and pathogen dependent cellular cytotoxicity [ADCC] are emerging as equally important.38,39 Ongoing investigations in murine models and patients increase the possibility that they may moreover stimulate adaptive immune responses in some settings.40
Immune Checkpoint Therapy:
The myriad of genetic and epigenetic alterations which are foible of all cancers provide a diverse set of antigens that the immune system can use to distinguish tumor cells from their normal counterparts. In the specimen of T cells, the ultimate width and quality of the response, which is initiated through antigen recognition by the TCR, is regulated by a wastefulness between co-stimulatory and inhibitory signals, that is immune checkpoints.41,42 Ipilimumab binds and blocks inhibitor signaling mediated by the T lamina surface coinhibitory molecule cytotoxic T lymphocyte antigen 4 [CTLA-4]. Considering the mechanism of whoopee is not specific to one tumor type, and considering a wealth of preclinical data support the role of tumor immune surveillance acros multiple malignancies43,44 Ipilimumab I stuff investigated as a treatment for patient with prostate, lung, renal, and breast cancer among other tumor types. 45 These drugs represent a radical and disruptive transpiration in cancer therapy in two ways. First, they target molecules involved in T lamina regulation as the soldiers of the immune system, rather than the tumor cell. Second, perhaps in a increasingly radical shift, the therapy is not designated to vivify the immune system to wade particular targets on tumor cells, but to remove inhibitory pathways that woodcut constructive antitumor T lamina responses. Understanding of immune checkpoint therapy has led to new weapons versus cancer which is elicit durable clinical responses and showed long-term remission for patiets, and provide an important whop in clinical advances well-nigh regulatory pathways in T cells and enhancing antitumor immune responses: Tumor cells express tumor specific antigens in the form of complexes of tumor-derived peptides unseat to MIIC molecules on the cell, this will be the target of T cells in this therapy. Tumor antigens can be derived from oncogenic viruses, differentiation antigens, epigenetically regulated molecules such as cancer testis antigens, or neoantigens derived from mutations associated with the process of carcinogenesis.46 Expression of B7 molecules is limited to subsets of hematopoietic cells, expressly DC, which have specialized the processes for efficient antigen presentation47 two laboratories independently showed that it oppose CD28 co stimulation and down-regulated T lamina responses48,49. Thus, vivification of T cells result in induction of expression of CTLA-4, which accumulates in the T lamina at the T lamina APC interface reaching a level where it sooner woodcut co-stimulation and abrogates an zingy T lamina response.
Molecular understructure of Cancer immunotherapy:
The human immune system mounts natural endogenous response to foreign cells, particularly highly immunogenic cancer cells, through a ramified series of steps. These involve presenting of cancer antigens to T-cells via antigen-presenting cells [APCs], priming and vitalizing T-cells in lymph nodes trafficking and infiltration of T-cells into tumor beds [tumor infiltrating lymphocytes], recognition of cancer cells by T cells, minutiae of antigen-specific systemic effector and memory T-cells, and humoral immunity, permitting effector T cells, other endogenous immune cells and antibodies to tumor to act in concert in order to eliminate cancer cells]. Both weakly and strongly immunogenic antigens on cancer cells enable multiple evasion strategies. This is suspected to be due to modulating factors in the tumor microenvironment that subvert the existing.50
Adoptive Immunotherapy:
T cells are moreover capable of sensing a variety of signals that can zestful them to potentially threatening pathogens and to cancer. Tumor specific T cells are probably zingy through encounters with tumor-associated antigens that are presented by specialized APC, including DC. However, zingy T cells are capable of directly recognizing antigens which were presented on the surfaces of tumor cells. Based on intravital imaging, there is a growing soul of vestige showing that the migration of tumor-specific T cells is rapidly underdeveloped when they encounter their cognate antigens.51 provide a favorable microenvironment that largest supports antitumor immunity. ACT is a living treatment considering the administered cells can proliferate in vivo and maintain their antitumor effector functions.52 Expanded TILs have cytolytic worriedness versus the original tumor and in unrelatedness to lymphokine zingy killer [LAK] cells the killing is MHC matriculation I restricted. They are selectively broaden from either tumor or draining lymph node cells via IL-2, and then re-stimulated with irradiated or killed tumor cells to maintain T lamina specificity.53
The topics to use ACT was facilitated by the unravelment of T lamina growth factor IL-2 in 1976, which provided a ways to grow T lymphocytes ex vivo, often without loss of effector functions.54 The uncontrived wardship of upper doses of IL-2 could inhibit tumor growth in mice55 and studies in 1982 demonstrated that the intravenous injection of immune lymphocytes expanded in IL-2 could powerfully treat unwieldy subcutaneous virus-induced lymphoma cells [FBL3]56. Moreover, wardship of IL-2 without lamina transfer could enhance therapeutic potential of these adoptively transferred lymphocytes.57 Expanded and zingy γδ T cells can mediate killing of glioblastoma and reduce tumor progression in mouse models.58
Cancer Vaccine therapy:
DC are designated as professional APC because of their capacity to provide T cells with all the signals required for antigen-specific T cell activation.59 The first signal is delivered to the TCR by MHC molecules presenting antigen-derived peptides. The second signal was provided by the binding of co-stimulatory molecules with their respective ligands on T-cells. A crucial positive co-stimulatory signal is provided by the interaction of the B7 family ligands CD80 and CD86, expressed by the DC, with the CD28 receptor expressed on the T cell surface. The third signal, that called polarization signal, determines the commitment of naive CD4+ T helper [TH0] cell towards TH1, TH2 or other fates. In the setting of cancer immunotherapy, the induction of a TH1 [cellular] immune response is highly desirable as this enables the generation of CTL capable of recognizing and destroying tumor cells in an antigen-specific fashion.60 NK cells have both cytotoxic and immunoregulatory functions and require priming to accomplish their full effector potential. Many of the cytokines that play a principal role in stimulating NK cell functions can be provided by DC, e.g., IL-12, IL-15, IL-18 and type I IFN.61 A more detailed understanding of the mechanisms leading to strong cellular immunity is necessary to enable rational approaches to the vaccine design. Two recent conceptual breakthroughs in this regard are our understanding that DC play a pivotal role in initiating the immune response to foreign antigens and the realizationthat adjuvants act primarily because they are DC activators.62
Therapeutic vaccines in chronic infections [or cancer] have two objectives: one is priming, whereas the other one is the modulation or reprogramming of memory cells, i.e., to transition from one type of immunity to another [e.g., regulatory to cytotoxic]. Tumor cells themselves are poor APC, which raises the question of how such potent immunity can be generated. Mouse models demonstrate that the generation of protective anti-tumor immunity relies on the presentation of tumor antigens by DC.63,64 An alternative to infusion of preformed tumor-specific antibodies or T cells, known as passive immunotherapy, is active specific immunotherapy [i.e., cancer vaccines] designed to elicit or boost similar tumor antibodies and T cells in patients. Some examples are vaccines against breast cancer [the HER2 antigen]65-66, B-cell lymphoma [the tumor immunoglobulin idiotype]67, lung cancer [the Mucin 1 cell surface associated [MUC1] antigen]68, melanoma [DC loaded with tumor peptides or killed tumor cells] 69,70, pancreatic cancer [telomerase peptides]71, and prostate cancer [DC loaded with prostatic acid phosphatase]72. The results of these trials are promising because in each there was evidence of an immune response to the vaccine, and in a few cases there were clinical responses with minimal or no adverse effects. Regarding the limited number of completed phase 3 trials, most have failed to present a significant benefit with respect to predetermined end points73.
CONCLUSION:
This paper we conclude that cancer immunology and immunotherapy are studied. We now have detailed knowledge of the molecular basis of cancer to allow a more personalized treatment based on genomic sequencing of an individual’s cancer cells to identify specific mutations in genes. These mutations can then be targeted with compounds to blockade the downstream pathways which drive cancer development and progression. Thus, each specific mutation serves as the predictive biomarker for selecting patients for treatment with a given agent. Clinical data generated principally over the past 5 years offer that we are at the threshold of golden era for adoptive T cell therapy, where advances in basic immunology have informed the development of a new field of synthetic immunology which may increase the potency.
REFERENCE:
1. Gravitz L. Cancer Immunotherapy. Nature. 2013; 504: S1 2. Palucka K. Q&A: Evidence presenter. Interview by Marian Turner. Nature. 2013; 504: S9. 60
2. Palucka K. Q&A: Evidence presenter. Interview by Marian Turner. Nature. 2013; 504: S9
3. Humphries C. Adoptive cell therapy: Honing that killer instinct.Nature. 2013; 504: S13-5.
4. Weintraub K. Drug development: Releasing the brakes. Nature. 2013; 504: S6-8
5. Littman DR. Releasing the brakes on cancer immunotherapy. Cell. 2015; 162: 1186-90.
6. Hodi FS, O’Day SJ, Mc Dermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010; 363: 711-23.
7. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015; 36: 265-76upon programmed cell death. EMBO J. 1992; 11: 3887-95.
8. Fialkow PJ. Clonal origin of human tumors. Biochim Biophys Acta. 1976; 458: 283-321.
9. Fidler IJ, Kripke ML. Metastasis results from preexisting variant cells within a malignant tumor. Science. 1977; 197: 893-5.
10. Nowell PC. The clonal evolution of tumor cell populations. Science. 1976; 194: 23-8.
11. Mueller MM, Fusenig NE. Friends or foes — bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004; 4: 839-49.
12. Jordan CT, Guzman ML, Noble M. Cancer stem cells. N Engl J Med. 2006; 355: 1253-61.
13. Cohnheim J. Congenitales, quergestreiftes Muskelsarkom der Nieren. Path Anat Physiol Klin Med. 1875; 65: 64-9.
14. Wicha MS, Liu S, Dontu G. Cancer stem cells: an old idea — a paradigm shift. Cancer Res. 2006; 66: 1883-90.
15. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997; 3: 730-7.
16. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994; 367: 645-8.
17. Yamashita T, Wang XW. Cancer stem cells in the development of liver cancer. J Clin Invest. 2013; 123: 1911-8.
18. Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004; 304: 1497-500.
19. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004; 350: 2129-39. Markowitz SD, Bertagnolli MM. Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 2009; 361: 2449-60.
20. Ogino S, Goel A. Molecular classification and correlates in colorectal cancer. J Mol Diagn. 2008; 10: 13-27.
21. Pritchard CC, Grady WM. Colorectal cancer molecular biology moves into clinical practice. Gut. 2011; 60: 116-29.
22. Dolle JM, Daling JR, White E, Brinton LA, Doody DR, Porter PL, et al. Risk factors for triple-negative breast cancer in women under the age of 45 years. Cancer Epidemiol Biomarkers Prev. 2009; 18: 1157-66.
23. Trivers KF, Lund MJ, Porter PL, Liff JM, Flagg EW, Coates RJ, et al. The epidemiology of triple-negative breast cancer, including race. Cancer Causes Control. 2009; 20: 1071-82.
24. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005; 310: 644-8.
25. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004; 304: 554.
26. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004; 304: 554.
27. Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007; 318: 1108-13.
28. Esteller M. Epigenetics in cancer. N Engl J Med. 2008; 358: 1148-59.
29. Piccart-Gebhart MJ, Procter M, Ley land-Jones B, Goldhirsch A, Untch M, Smith I, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005; 353: 1659-72.
30. Mendelsohn J. Epidermal growth factor receptor inhibition by a monoclonal antibody as anticancer therapy. Clin Cancer Res. 1997; 3: 2703-7.
31. Giaccone G. Epidermal growth factor receptor inhibitors in the treatment of non-small-cell lung cancer. J Clin Oncol. 2005; 23: 3235-42.
32. Kim ES, Vokes EE, Kies MS. Cetuximab in cancers of the lung and head & neck. Semin Oncol. 2004; 31 [Suppl 1]: 61-7.
33. Finn OJ. Cancer Immunology. N Engl J Med. 2008; 358: 2704-15.
34. Melero I, Hervas-Stubbs S, Glennie M, Pardoll DM, Chen L. Immunostimulatory monoclonal antibodies for cancer therapy. Nat Rev Cancer. 2007; 7: 95-106.
35. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab [anti-PD-1] in melanoma. N Engl J Med. 2013: 369: 134-44.
36. Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015; 33: 1889-94.
37. Jiang XR, Song A, Bergelson S, Arroll T, Parekh B, May K, et al. Advances in the assessment and control of the effector functions of therapeutic antibodies. Nat Rev Drug Discov. 2011; 10: 101-11.
38. Weiner LM, Murray JC, Shuptrine CW. Antibody-based immunotherapy of cancer. Cell. 2012; 148: 1081-4.
39. Taylor C, Hershman D, Shah N, Suciu-Foca N, Petrylak DP, Taub R, et al. Augmented HER-2 specific immunity during treatment with trastuzumab and chemotherapy. Clin Cancer Res. 2007; 13: 5133-43
40. Zou W, Chen L. Inhibitory B7-family molecules in the tumour microenvironment. Nature Rev Immunol. 2008; 8: 467-77.
41. Andre F, Dieci MV, Dubsky P, Sotiriou C, Curigliano G, Denkert C, et al. Molecular pathways: involvement of immune pathways in the therapeutic response and outcome in breast cancer. Clin Cancer Res. 2013; 19: 28-33.
42. May KF Jr, Gulley JL, Drake CG, Dranoff G, Kantoff PW. Prostate cancer immunotherapy. Clin Cancer Res. 2011; 17: 5233-8.
43. Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med. 2014; 65: 185-202.
44. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015; 348: 69-74.
45. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015 ; 348: 56-61.
46. Walunas TL, Lenschow DJ, Bakker CY, Linsley PS, Freeman GJ, Green JM, et al. CTLA-4 can function as a negative regulator of T cell activation. Immunity. 1994; 1: 405-13.
47. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J Exp Med. 1995; 182: 459-65
48. Deguine J, Breart B, Lemaitre F, Di Santo JP, Bousso P. Intravital imaging reveals distinct dynamics for natural killer and CD8+ T cells during tumor regression. Immunity. 2010; 33: 632-44.
49. Restivo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012; 12: 269-81.
50. Ruf P, Lindhofer H. Induction of a long-lasting antitumor immunity by a trifunctional bispecific antibody. Blood. 2001; 98: 2526-34.
51. Morgan DA, Ruscetti FW, Gallo R. Selective in vitro growth of T lymphocytes from normal human bone marrows. Science. 1976; 193: 1007-8.
52. Rosenberg SA, Mulé JJ, Spiess PJ, Reichert CM, Schwarz SL. Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2. J Exp Med. 1985; 161: 1169-88.
53. Eberlein TJ, Rosenstein M, Rosenberg SA. Regression of a disseminated syngeneic solid tumor by systemic transfer of lymphoid cells expanded in interleukin 2. J Exp Med. 1982; 156: 385-97.
54. 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. J Immunol. 1984; 132: 2123-8.
55. Bryant NL, Gillespie GY, Lopez RD, Markert JM, Cloud GA, Langford CP, et al. Preclinical evaluation of ex vivo expanded/activated gammadelta T cells for immunotherapy of glioblastoma multiforme. J Neurooncol. 2011; 101: 179-88.
56. Cools N, Ponsaerts P, Van Tendeloo VF, Berneman ZN. Balancing between immunity and tolerance: an interplay between dendritic cells, regulatory T cells, and effector T cells. J Leukoc Biol. 2007; 82: 1365-74.
57. Zinkernagel RM, Hengartner H. Regulation of the immune response by antigen. Science. 2001; 293: 251-3.
58. Reise Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006; 6: 476-83.
59. Anguile S, Willemen Y, Lion E, Smits EL, Berneman ZN. Dendritic cell vaccination in acute myeloid leukemia. Cytotherapy. 2012; 14: 647-56.
60. Moretta L, Ferlazzo G, Bottino C, Vitale M, Pende D, Mingari MC, et al. Effector and regulatory events during natural killer-dendritic cell interactions. Immunol Rev. 2006; 214: 219-28.
61. Palucka K, Banchereau J, Mellman I. Designing vaccines based on biology of human dendritic cell subsets. Immunity. 2010; 33: 464-78.
62. Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn GP, Archambault JM, et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J Exp Med. 2011; 208: 1989-2003.
63. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy KM, et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J Exp Med. 2011; 208: 2005-16.
64. Disis ML, Schiffman K, Guthrie K, Salazar LG, Knutson KL, Goodell V, et al. Effect of dose on immune response in patients vaccinated with an her-2/neu intracellular domain protein-based vaccine. J Clin Oncol. 2004; 22: 1916-25.
65. Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R, et al. Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res. 2007; 67: 1842-52.
66. Baxevanis CN, Sotiriadou NN, Gritzapis AD, Sotiropoulou PA, Perez SA, Cacoullos NT, et al. Immunogenic HER-2/neu peptides as tumor vaccines. Cancer Immunol Immunother. 2006; 55: 85-95.
67. Redfern CH, Guthrie TH, Bessudo A, Densmore JJ, Holman PR, Janakiraman N, et al. Phase II trial of idiotype vaccination in previously treated patients with indolent non-Hodgkin’s lymphoma resulting in durable clinical responses. J Clin Oncol. 2006; 24: 3107-12.
68. Butts C, Murray N, Maksymiuk A, Goss G, Marshall E, Soulières D, et al. Randomized phase IIB trial of BLP25 liposome vaccine in stage IIIB and IV non-small-cell lung cancer. J Clin Oncol. 2005; 23: 6674-81.
69. Fay JW, Palucka AK, Paczesny S, Dhodapkar M, Johnston DA, Burkeholder S, et al. Long-term outcomes in patients with metastatic melanoma vaccinated with melanoma peptide-pulsed CD34[+] progenitor-derived dendritic cells. Cancer Immunol Immunother. 2006; 55: 1209-18.
70. Palucka AK, Ueno H, Connolly J, Kerneis-Norvell F, Blanck JP, Johnston DA, et al. Dendritic cells loaded with killed allogeneic melanoma cells can induce objective clinical responses and MART-1 specific CD8+ T-cell immunity. J Immunother. 2006; 29: 545-57.
71. Bernhardt SL, Gjertsen MK, Trachsel S, Moller M, Eriksen JA, Meo M, et al. Telomerase peptide vaccination of patients with nonresectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer. 2006; 95: 1474-82.
72. Gould P. Sipuleucel-T shows partial advantage in prostate cancer. Lancet Oncol. 2006; 7: 710.
73. Finke LH, Wentworth K, Blumenstein B, Rudolph NS, Levitsky H, Hoos A. Lessons from randomized phase III studies with active cancer immunotherapies — outcomes from the 2006 meeting of the Cancer Vaccine Consortium [CVC]. Vaccine. 2007; 25 [Suppl 2]: B97-109.
Received on 23.05.2022 Modified on 03.06.2022
Accepted on 12.06.2022 ©A&V Publications All right reserved
Res. J. Pharmacology and Pharmacodynamics.2022;14(3):159-164.
DOI: 10.52711/2321-5836.2022.00027