Skip to main content

Oncogene withdrawal engages the immune system to induce sustained cancer regression

Abstract

The targeted inactivation of a single oncogene can induce dramatic tumor regression, suggesting that cancers are “oncogene addicted.” Tumor regression following oncogene inactivation has been thought to be a consequence of restoration of normal physiological programs that induce proliferative arrest, apoptosis, differentiation, and cellular senescence. However, recent observations illustrate that oncogene addiction is highly dependent upon the host immune cells. In particular, CD4+ helper T cells were shown to be essential to the mechanism by which MYC or BCR-ABL inactivation elicits “oncogene withdrawal.” Hence, immune mediators contribute in multiple ways to the pathogenesis, prevention, and treatment of cancer, including mechanisms of tumor initiation, progression, and surveillance, but also oncogene inactivation-mediated tumor regression. Data from both the bench and the bedside illustrates that the inactivation of a driver oncogene can induce activation of the immune system that appears to be essential for sustained tumor regression.

Introduction

Capitalizing on oncogene addiction: a therapeutic objective

The inactivation of a single oncogene can result in the dramatic and sustained regression of some cancers [14]. Targeted inactivation of an oncogene can be associated with proliferative arrest, apoptosis and/or senescence, and differentiation [3]. Oncogene addiction appears to be a consequence of the restoration of physiological programs [2, 5], but also has been described as a consequence of synthetic lethality [6] and the differential decay of survival and apoptosis programs [7]. “Oncogene withdrawal” occurs upon suppression of initiating genetic events in tumors [8, 9]. It is not known when a cancer will be addicted to a particular oncogene [4]. Oncogene addiction has been thought to occur through host cell autonomous, tumor intrinsic mechanisms. Yet, recent observations illustrate that oncogene addiction has both cell autonomous as well as immune-mediated mechanisms [1014] (Figure 1).

Figure 1
figure 1

The host immune system is required for sustained tumor regression following oncogene withdrawal. Following oncogene inactivation in a mouse model by transgenic methods or in patients by oncogene-targeted therapy, there are tumor cell-intrinsic consequences, immunological consequences, and host microenvironmental consequences. Tumor cell-intrinsic consequences include proliferative arrest and the induction of apoptosis. Dying tumor cells and antigen debris may stimulate an immune response, which may in turn feed back in to the tumor cell-intrinsic consequences. The immune response, particularly helper T cells, can influence environmental consequences, including the induction of senescence and the collapse of angiogenesis. Lastly, senescing tumor cells may have a secretory phenotype, which in turn may influence the immune system. Taken together, these three components lead to a remodeling of the entire tumor (both in the cancer cells and in the environment) and contribute to lasting tumor regression and protection from relapse.

Oncogene addiction has been largely studied in mouse models. However, tumor regression following oncogene inactivation has been observed in response to targeted therapeutics in humans including molecules that target BCR-ABL or c-Kit, EGFR, ALK, BRAF V600E, PML-RARα, and HER2/neu for the treatment of leukemia, lung adenocarcinoma, non-small cell lung cancer, melanoma, and breast cancer [1523]. Other drugs are in clinical investigation, including those that target JAK2, MDM2, and PI3 Kinase [2431]; drugs that target RAS [32] and MYC [33, 34] are in early development. It remains to be seen if these agents specifically take advantage of oncogene addiction. In general, little has been done to study the mechanism of action of these therapeutic agents in human patients.

Experimental mouse models have been a particularly tractable approach to interrogating the mechanism of oncogene addiction. Transgenic mouse models employing strategies that enable the conditional expression of oncogenes have been used to illustrate that cancers initiated by an oncogene, such as MYC, RAS, BCR-ABL, MET, and BRAF, are reversible upon suppression of the oncogene [1, 3540].

Review

Cancer and the immune system: a complex relationship

The mechanisms by which targeted therapies engage oncogene addiction have been presumed to be cell autonomous. However, oncogene inactivation causes dramatic changes in the microenvironment including the shut down of angiogenesis [4144] and the recruitment of host effector cells, including innate and adaptive immune cells [4, 4548]. Thus it appeared likely that the immune system is playing an active role in the mechanism of tumor regression following oncogene inactivation.

Furthermore, it is well known that the immune system is a barrier to tumorigenesis [49]. Hosts with absent or suppressed immune systems have a greatly increased incidence of many different types of cancer [50, 51]. Patients who are transplant recipients and take drugs that suppress their adaptive immunity demonstrate dramatically increased incidences of lymphoma and squamous cell carcinoma [5254]. Moreover, patients who are immunosuppressed also have an impeded response to cancer therapy with a decreased overall and progression-free survival [55, 56]. Thus, immune surveillance mechanisms are critical both to the prevention as well the efficacy of conventional treatment of these cancers [5759] and are a critical component to the therapeutic efficacy of agents for cancer [54, 6062].

Correspondingly, the activation of the immune system through specific immune-based therapies is efficacious for the treatment of some cancers. This includes antibodies that target cancer cells, such as Rituximab [63] and Trastuzumab [64], as well as antibodies or drugs that modulate immunostimulatory or immunoinhibitory signals [6567], such as anti-CTLA-4 [68] and anti-PD-L1 [69]. The combination of conventional chemotherapy with targeted immune therapy has emerged as an effective approach for the treatment of some cancers.

Oncogene inactivation activates the immune system

Recent studies in experimental mouse models illustrate the mechanisms by which oncogene “withdrawal” results in immune activation (Figure 1, [24, 45]). In a tetracycline-regulated conditional mouse model of MYC-induced T cell Acute Lymphoblastic Leukemia (T-ALL), the tumor cells undergo proliferative arrest and death within 2 days of turning off the MYC oncogene via tumor intrinsic, host independent, immune independent mechanisms. Subsequently, between 2 and 5 days, there is a recruitment of immune effector cells that are required to induce cellular senescence of tumor cells and the shut down of angiogenesis in the tumor microenvironment [70]. The kinetics of tumor regression, the extent of tumor regression, and the ability to maintain sustained tumor regression are all compromised in immunodeficient hosts.

Provocatively, CD4+ helper T cells were found to be the key immune effector required for oncogene inactivation-induced tumor regression in the conditional MYC-driven T-ALL mouse model. The CD4+ T cells are likely to contribute to tumor regression through many mechanisms. Of note, CD4+ T cells can express a variety of cytokines that have been implicated in the regulation of cellular senescence and/or angiogenesis [7174]. CD4+ T cells may also be working via direct cellular interactions with the tumor cells or host stromal cells in the tumor microenvironment. Finally, CD4+ T cells appear to recruit other immune and host cells.

The CD4+ helper T cells must express thrombospondins in order to contribute to tumor regression following oncogene inactivation [45]. TSP-1 has been suggested to be a key regulator of both angiogenesis and senescence [75]. Moreover, CD47, the receptor of TSP-1, is a key regulator of the immune response [76]. TSP-1 and CD47 have been suggested to regulate cellular senescence [75, 77, 78]. However, there is also a general induction of anti-tumor and a suppression of pro-tumor cytokines after oncogene inactivation that occurs only in immunocompetent hosts [45]. Hence, specific secreted factors are likely to contribute to the mechanism of oncogene addiction and withdrawal.

How oncogene inactivation recruits a response of CD4+ T cells is not known. There are several possibilities. First, oncogenes such as MYC have been suggested to regulate the expression of molecules that may be immunosuppressive and/or regulate angiogenesis. Hence, MYC inactivation could lead to the direct change in expression of cytokines by tumor cells, thereby recruiting immune cells [79]. Second, oncogene inactivation could activate an immune response through immunogenic cell death that in turns activates the immune response [80]. Identifying the specific mechanism of the immune activation and response could suggest important strategies for monitoring and implementing a therapeutic response [10].

Importantly, many other immune effectors are likely to contribute to the response of targeted therapies. This is potentially governed by the unique genetic and cellular context of each tumor [81, 82]. In other mouse models, investigators have noted that innate immune cells such as mast cells [83], macrophages [84], and other antigen-presenting cells (APCs) may function as barriers to tumor growth and facilitators of tumor regression. Thus, it is likely that these other innate and adaptive immune cells contribute to the mechanism of oncogene addiction and tumor regression following oncogene inactivation.

In the clinic: targeted oncogene inactivation and immune response

Oncogene addiction has been studied in a more limited manner in human patients. Some studies indicate that the host immune response is essential for the optimal response to conventional chemotherapy and radiotherapy [8587]. A major potential limitation of conventional therapeutics is that they often suppress the immune response [88].

Other correlative studies suggest that an immune response may contribute to the mechanism of targeted oncogene inactivation. In human patients with BCR-ABL+ gastrointestinal stromal tumors (GIST) treated with Imatinib, IFN-γ secretion by NK cells in the peripheral blood is associated with a better clinical response [89]. Similarly, the inhibition of BRAF both directly inhibits tumor growth but also appears to activate the immune system [90]. The combination of a BRAF inhibitor, Vemurafenib, with immune therapy may be more effective in the treatment of tumors [91]. Moreover, Vemurafenib was associated with intratumoral accumulation of adoptively transferred T cells [92] as well as increased intratumoral numbers of CD4+ or CD8+ T cells [90] and this was associated with a better prognosis [90]. Other studies have shown that BRAF inhibition is associated with the reduction of immunosuppressive cytokines and chemokines [93]. Ongoing clinical studies are examining if Vemurafenib in combination with immunotherapy is more clinically effective [94, 95].

Other targeted therapies may induce an immune response in addition to their tumor-specific effects. Sunitinib, which targets PDGFR, RET, and KIT, recruits an immune response that may contribute to its mechanism [96] through the induction of IFN-γ-producing T cells [97] and decreased regulatory T cells [97, 98]. Arsenic and all-trans-retinoic-acid (ATRA), used for the treatment of PML-RARα acute promylecytic leukemia, is associated with altered antigen presentation [99]. Bortezomib is a proteasome inhibitor used in the treatment of hematopoietic tumors and is associated with the recruitment to tumor sites of CD8+ T cells and dendritic cells [100]. The EGFR inhibitor, Erlotinib, is effective in the treatment of non-small cell lung cancer and is associated with increased intratumoral numbers of dendritic cells [101]. Trastuzumab targets HER2/neu for the treatment of breast and ovarian cancer and may require an NK cell response [102, 103]. Thus, targeted inhibition of oncogenes may be efficacious in part through the activation of an immune response.

In some cases, targeted inactivation of oncogenes could inhibit an immune response and impede the efficacy of an anti-tumor therapeutic. For example, inhibition of MAPK/extracellular signal-regulated kinase kinase (MEK) results in T cell inhibition [104]. Imatinib can affect the immune response in a multitude of ways [24, 45, 105109]. Thus, it will be pivotal to consider how targeted oncogene inactivation can induce or suppress an immune response and how this may contribute to the mechanism of action of anti-neoplastic agents.

Therapeutic implications for oncogene-targeted therapies

Experimental evidence and clinical observations suggest that targeted oncogene inactivation generates an anti-tumor immune response. More generally this suggests that targeted oncogene inactivation can be exploited as an immune therapy. Unlike conventional chemotherapy or radiotherapy, the judicious choice of agents that target specific oncogenes may lead to tumor regression both by directly targeting tumor cells and indirectly by inducing a robust immune response. If this were the case, it would have several practical implications for the development and application of therapeutics.

First, the combination of oncogene-targeted therapy with specific immunomodulatory therapy may further increase the clinical response and long-term survival of patients [94, 110, 111]. Pointedly, immune activation may be essential to prevent the emergence of therapy-resistant tumor cells, which can lead to tumor recurrence [112, 113]. Hence, the identification of the best agents to prompt oncogene withdrawal will require examination of the efficacy of these therapies with consideration of their ability to induce both cell autonomous and host-dependent mechanisms of tumor regression.

Several targeted therapies are currently approved or under investigation in combination with immunomodulatory therapies (Table 1). For the treatment of melanoma, MEK and VEGF inhibitors are being administered with Ipilimumab [114, 115] and IL-2 [116], respectively. BRAF inhibitors are being examined together with Ipilimumab [117]. Ipilimumab is also being interrogated in combination with Brentuximab for the treatment of Hodgkin’s Lymphoma [118] and with Crizotinib for non-small cell lung cancer [119]. Ipilimumab and anti-PD-L1 inhibitors are being analyzed in combination with Erlotinib in non-small cell lung cancer [119, 120].

Targeted therapies together with immune-based therapies are also being examined for the treatment of other types of cancer. Lenalidomide and Bortezomib are being examined for treatment of multiple myeloma [121], Lenalidomide and Ibrutinib are under investigation for Chronic Lymphocytic Leukemia [122], and Nivolumab is being administered with Sunitinib for renal cell cancer [123]. Additionally, the mTOR inhibitor Temsirolimus is being studied with Interferon-α for renal cancer [124]. Imatinib and Rituximab are being investigated in combination with Nivolumab [125] or Pidilizumab [126]. Trastuzumab is under investigation with peptide vaccines and cytokines [127]. These investigations may identify combinations of targeted and immune-based therapies that are more efficacious for the treatment of cancer. Furthermore, the appreciation that immune activation may be a critical component to the efficacy of therapeutics may be important for the measurement and maximization of their clinical efficacy.

Table 1 Targeted therapies studied or under investigation in cooperation with immune therapies

Conclusions

Experimental and clinical observations suggest a model of oncogene addiction and a role for the immune system (Figure 1). The inactivation of an oncogene in a tumor appears to initiate cancer cell-intrinsic programs of tumor regression including proliferative arrest, differentiation, and apoptosis, as well as immune-dependent modulation of the microenvironment that contributes to cellular senescence and the shut down of angiogenesis. These mechanisms are collectively required for complete and sustained tumor regression.

Oncogene inactivation in a tumor results in activation of an immune response (Figure 1). The mechanisms by which this occurs are not defined. These mechanisms potentially involve both direct mechanisms related to the production of immune recruiting cytokines as well as more indirect mechanisms such as immunogenic cell death. Many cellular and cytokine effectors are likely to be involved, including CD4+ T cells, CD8+ T cells, B cells, and innate immune cells such as macrophages and NK cells (Figure 1). It is possible that the impairment of specific cellular, humoral, or chemokine mechanisms would facilitate the re-emergence of tumor cells that are refractory to targeted therapy.

There are several practical implications of this model. First, successful targeted therapy against a cancer is likely to require an intact host immune system. Second, the measurement of the efficacy of a targeted therapy is likely to be most readily defined through interrogation of immune activation after drug administration. Third, the early development of therapeutic agents should be performed using model systems that have an intact host immune system as opposed to in vitro model systems or xenograft model systems in severely immunocompromised animals.

Our model predicts that the immune system not only directly eliminates tumor cells but also plays a critical role in modulating the tumor microenvironment. Diagnostic assays that detect an immune response may predict the therapeutic efficacy of oncogene-targeted agents. Strategies need to be developed that would enable the measurement of these effector cells and molecules before and after therapeutic treatment. This could include in situ measurements in patients using flow cytometry analysis of immune effector cells, proteomic and genomic analysis, and noninvasive molecular imaging methods.

Finally, the most effective clinical strategy to treat tumors will likely require a coordination of therapies that target oncogenes in combination with the activation of specific immune effectors. Conversely, existing conventional chemotherapies that often impede an immune response may antagonize the efficacy of targeted therapeutics. Hence, mechanistic insight into how oncogene withdrawal prompts immune activation may actualize rationale therapeutic strategies.

References

  1. Felsher DW, Bishop JM: Reversible tumorigenesis by MYC in hematopoietic lineages. Mol Cell. 1999, 4: 199-207.

    CAS  PubMed  Google Scholar 

  2. Felsher DW: Cancer revoked: oncogenes as therapeutic targets. Nat Rev Cancer. 2003, 3: 375-380.

    CAS  PubMed  Google Scholar 

  3. Weinstein IB: Cancer. Addiction to oncogenes--the Achilles heal of cancer. Science. 2002, 297: 63-64.

    CAS  PubMed  Google Scholar 

  4. Hanahan D, Weinberg RA: Hallmarks of cancer: the next generation. Cell. 2011, 144: 646-674.

    CAS  PubMed  Google Scholar 

  5. Felsher DW: Oncogene addiction versus oncogene amnesia: perhaps more than just a bad habit?. Cancer Res. 2008, 68: 3081-3086. discussion 3086

    CAS  PubMed  Google Scholar 

  6. Kaelin WG: The concept of synthetic lethality in the context of anticancer therapy. Nat Rev Cancer. 2005, 5: 689-698.

    CAS  PubMed  Google Scholar 

  7. Sharma SV, Settleman J: Oncogene addiction: setting the stage for molecularly targeted cancer therapy. Genes Dev. 2007, 21: 3214-3231.

    CAS  PubMed  Google Scholar 

  8. Bozic I, Antal T, Ohtsuki H, Carter H, Kim D, Chen S, Karchin R, Kinzler KW, Vogelstein B, Nowak MA: Accumulation of driver and passenger mutations during tumor progression. Proc Natl Acad Sci U S A. 2010, 107: 18545-18550.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wood LD, Parsons DW, Jones S, Lin J, Sjoblom T, Leary RJ, Shen D, Boca SM, Barber T, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y, Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipitsin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV: The genomic landscapes of human breast and colorectal cancers. Science. 2007, 318: 1108-1113.

    CAS  PubMed  Google Scholar 

  10. Restifo NP: Can antitumor immunity help to explain "oncogene addiction"?. Cancer Cell. 2010, 18: 403-405.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Furth PA: Cancer prevention as biomodulation: targeting the initiating stimulus and secondary adaptations. Ann N Y Acad Sci. 2012, 1271: 1-9.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ding ZC, Huang L, Blazar BR, Yagita H, Mellor AL, Munn DH, Zhou G: Polyfunctional CD4(+) T cells are essential for eradicating advanced B-cell lymphoma after chemotherapy. Blood. 2012, 120: 2229-2239.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ding ZC, Zhou G: Cytotoxic chemotherapy and CD4+ effector T cells: an emerging alliance for durable antitumor effects. Clin Dev Immunol. 2012, 2012: 890178-

    PubMed  PubMed Central  Google Scholar 

  14. Anders K, Buschow C, Herrmann A, Milojkovic A, Loddenkemper C, Kammertoens T, Daniel P, Yu H, Charo J, Blankenstein T: Oncogene-targeting T cells reject large tumors while oncogene inactivation selects escape variants in mouse models of cancer. Cancer Cell. 2011, 20: 755-767.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Shortt J, Johnstone RW: Oncogenes in cell survival and cell death. Cold Spring Harb Perspect Biol. 2012, 1: 4(12)-

    Google Scholar 

  16. Baker SJ, Reddy EP: Targeted inhibition of kinases in cancer therapy. Mt Sinai J Med. 2010, 77: 573-586.

    PubMed  PubMed Central  Google Scholar 

  17. Houshmand P, Zlotnik A: Targeting tumor cells. Curr Opin Cell Biol. 2003, 15: 640-644.

    CAS  PubMed  Google Scholar 

  18. Bollag G, Tsai J, Zhang J, Zhang C, Ibrahim P, Nolop K, Hirth P: Vemurafenib: the first drug approved for BRAF-mutant cancer. Nat Rev Drug Discov. 2012, 11: 873-886.

    CAS  PubMed  Google Scholar 

  19. O'Bryant CL, Wenger SD, Kim M, Thompson LA: Crizotinib: a new treatment option for ALK-positive non-small cell lung cancer. Ann Pharmacother. 2013, 47: 189-197.

    PubMed  Google Scholar 

  20. Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C: ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC. Target Oncol. 2013, 8: 55-67.

    PubMed  Google Scholar 

  21. Blay JY, Le Cesne A, Alberti L, Ray-Coquart I: Targeted cancer therapies. Bull Cancer. 2005, 92: E13-18.

    PubMed  Google Scholar 

  22. Soria JC, Blay JY, Spano JP, Pivot X, Coscas Y, Khayat D: Added value of molecular targeted agents in oncology. Ann Oncol. 2011, 22: 1703-1716.

    CAS  PubMed  Google Scholar 

  23. Nagai S, Takahashi T, Kurokawa M: The impact of molecularly targeted therapies upon the understanding of leukemogenesis and the role of hematopoietic stem cell transplantation in acute promyelocytic leukemia. Curr Stem Cell Res Ther. 2010, 5: 372-378.

    CAS  PubMed  Google Scholar 

  24. Casey SC, Bellovin DI, Felsher DW: Noncanonical roles of the immune system in eliciting oncogene addiction. Curr Opin Immunol. 2013, 25: 246-258.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Lindeman NI, Cagle PT, Beasley MB, Chitale DA, Dacic S, Giaccone G, Jenkins RB, Kwiatkowski DJ, Saldivar JS, Squire J, Thunnissen E, Ladanyi M: Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. Arch Pathol Lab Med. 2013, 137: 828-860.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Younes A, Romaguera J, Fanale M, McLaughlin P, Hagemeister F, Copeland A, Neelapu S, Kwak L, Shah J, de Castro Faria S, Hart S, Wood J, Jayaraman R, Ethirajulu K, Zhu J: Phase I study of a novel oral Janus kinase 2 inhibitor, SB1518, in patients with relapsed lymphoma: evidence of clinical and biologic activity in multiple lymphoma subtypes. J Clin Oncol. 2012, 30: 4161-4167.

    CAS  PubMed  Google Scholar 

  27. Cheok CF, Verma CS, Baselga J, Lane DP: Translating p53 into the clinic. Nat Rev Clin Oncol. 2011, 8: 25-37.

    CAS  PubMed  Google Scholar 

  28. Essmann F, Schulze-Osthoff K: Translational approaches targeting the p53 pathway for anti-cancer therapy. Br J Pharmacol. 2012, 165: 328-344.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Fruman DA, Rommel C: PI3Kdelta inhibitors in cancer: rationale and serendipity merge in the clinic. Cancer Discov. 2011, 1: 562-572.

    CAS  PubMed  Google Scholar 

  30. Roschewski M, Farooqui M, Aue G, Wilhelm F, Wiestner A: Phase I study of ON 01910.Na (Rigosertib), a multikinase PI3K inhibitor in relapsed/refractory B-cell malignancies. Leukemia. 2013, 27: 1920-1923.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Hong DS, Bowles DW, Falchook GS, Messersmith WA, George GC, O'Bryant CL, Vo AC, Klucher K, Herbst RS, Eckhardt SG, Peterson S, Hausman DF, Kurzrock R, Jimeno A: A multicenter phase I trial of PX-866, an oral irreversible phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors. Clin Cancer Res. 2012, 18: 4173-4182.

    CAS  PubMed  Google Scholar 

  32. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM: K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013, 503: 548-551.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Ott CJ, Kopp N, Bird L, Paranal RM, Qi J, Bowman T, Rodig SJ, Kung AL, Bradner JE, Weinstock DM: BET bromodomain inhibition targets both c-Myc and IL7R in high-risk acute lymphoblastic leukemia. Blood. 2012, 120: 2843-2852.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Delmore JE, Issa GC, Lemieux ME, Rahl PB, Shi J, Jacobs HM, Kastritis E, Gilpatrick T, Paranal RM, Qi J, Chesi M, Schinzel AC, McKeown MR, Heffernan TP, Vakoc CR, Bergsagel PL, Ghobrial IM, Richardson PG, Young RA, Hahn WC, Anderson KC, Kung AL, Bradner JE, Mitsiades CS: BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011, 146: 904-917.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Huettner CS, Zhang P, Van Etten RA, Tenen DG: Reversibility of acute B-cell leukaemia induced by BCR-ABL1. Nat Genet. 2000, 24: 57-60.

    CAS  PubMed  Google Scholar 

  36. Chin L, Tam A, Pomerantz J, Wong M, Holash J, Bardeesy N, Shen Q, O'Hagan R, Pantginis J, Zhou H, Horner JW, Cordon-Cardo C, Yancopoulos GD, DePinho RA: Essential role for oncogenic Ras in tumour maintenance. Nature. 1999, 400: 468-472.

    CAS  PubMed  Google Scholar 

  37. Jain M, Arvanitis C, Chu K, Dewey W, Leonhardt E, Trinh M, Sundberg CD, Bishop JM, Felsher DW: Sustained loss of a neoplastic phenotype by brief inactivation of MYC. Science. 2002, 297: 102-104.

    CAS  PubMed  Google Scholar 

  38. Shachaf CM, Kopelman AM, Arvanitis C, Karlsson A, Beer S, Mandl S, Bachmann MH, Borowsky AD, Ruebner B, Cardiff RD, Yang Q, Bishop JM, Contag CH, Felsher DW: MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature. 2004, 431: 1112-1117.

    CAS  PubMed  Google Scholar 

  39. Hoeflich KP, Gray DC, Eby MT, Tien JY, Wong L, Bower J, Gogineni A, Zha J, Cole MJ, Stern HM, Murray LJ, Davis DP, Seshagiri S: Oncogenic BRAF is required for tumor growth and maintenance in melanoma models. Cancer Res. 2006, 66: 999-1006.

    CAS  PubMed  Google Scholar 

  40. Boxer RB, Jang JW, Sintasath L, Chodosh LA: Lack of sustained regression of c-MYC-induced mammary adenocarcinomas following brief or prolonged MYC inactivation. Cancer Cell. 2004, 6: 577-586.

    CAS  PubMed  Google Scholar 

  41. Giuriato S, Ryeom S, Fan AC, Bachireddy P, Lynch RC, Rioth MJ, van Riggelen J, Kopelman AM, Passegue E, Tang F, Folkman J, Felsher DW: Sustained regression of tumors upon MYC inactivation requires p53 or thrombospondin-1 to reverse the angiogenic switch. Proc Natl Acad Sci U S A. 2006, 103: 16266-16271.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Baudino TA, McKay C, Pendeville-Samain H, Nilsson JA, Maclean KH, White EL, Davis AC, Ihle JN, Cleveland JL: c-Myc is essential for vasculogenesis and angiogenesis during development and tumor progression. Genes Dev. 2002, 16: 2530-2543.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Brandvold KA, Neiman P, Ruddell A: Angiogenesis is an early event in the generation of myc-induced lymphomas. Oncogene. 2000, 19: 2780-2785.

    CAS  PubMed  Google Scholar 

  44. Janz A, Sevignani C, Kenyon K, Ngo CV, Thomas-Tikhonenko A: Activation of the myc oncoprotein leads to increased turnover of thrombospondin-1 mRNA. Nucleic Acids Res. 2000, 28: 2268-2275.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Rakhra K, Bachireddy P, Zabuawala T, Zeiser R, Xu L, Kopelman A, Fan AC, Yang Q, Braunstein L, Crosby E, Ryeom S, Felsher DW: CD4(+) T cells contribute to the remodeling of the microenvironment required for sustained tumor regression upon oncogene inactivation. Cancer Cell. 2010, 18: 485-498.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Albini A, Sporn MB: The tumour microenvironment as a target for chemoprevention. Nat Rev Cancer. 2007, 7: 139-147.

    CAS  PubMed  Google Scholar 

  47. Coussens LM, Tinkle CL, Hanahan D, Werb Z: MMP-9 supplied by bone marrow-derived cells contributes to skin carcinogenesis. Cell. 2000, 103: 481-490.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Bissell MJ, Radisky D: Putting tumours in context. Nat Rev Cancer. 2001, 1: 46-54.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Gasser S, Raulet D: The DNA damage response, immunity and cancer. Semin Cancer Biol. 2006, 16: 344-347.

    CAS  PubMed  Google Scholar 

  50. Chaturvedi AK, Pfeiffer RM, Chang L, Goedert JJ, Biggar RJ, Engels EA: Elevated risk of lung cancer among people with AIDS. AIDS. 2007, 21: 207-213.

    PubMed  Google Scholar 

  51. Mbulaiteye SM, Biggar RJ, Goedert JJ, Engels EA: Immune deficiency and risk for malignancy among persons with AIDS. J Acquir Immune Defic Syndr. 2003, 32: 527-533.

    PubMed  Google Scholar 

  52. Dugue PA, Rebolj M, Garred P, Lynge E: Immunosuppression and risk of cervical cancer. Expert Rev Anticancer Ther. 2013, 13: 29-42.

    CAS  PubMed  Google Scholar 

  53. Kubica AW, Brewer JD: Melanoma in immunosuppressed patients. Mayo Clin Proc. 2012, 87: 991-1003.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoover RN: Lymphoma risks in populations with altered immunity--a search for mechanism. Cancer Res. 1992, 52: 5477s-5478s.

    CAS  PubMed  Google Scholar 

  55. Boshoff C, Weiss R: AIDS-related malignancies. Nat Rev Cancer. 2002, 2: 373-382.

    CAS  PubMed  Google Scholar 

  56. Ray-Coquard I, Cropet C, Van Glabbeke M, Sebban C, Le Cesne A, Judson I, Tredan O, Verweij J, Biron P, Labidi I, Guastalla JP, Bachelot T, Perol D, Chabaud S, Hogendoorn PC, Cassier P, Dufresne A, Blay JY: Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009, 69: 5383-5391.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Al-Tameemi M, Chaplain M, d'Onofrio A: Evasion of tumours from the control of the immune system: consequences of brief encounters. Biol Direct. 2012, 7: 31-

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ribas A: Immunoediting the cancer genome–a new approach for personalized cancer therapy?. Pigment Cell Melanoma Res. 2012, 25: 297-298.

    PubMed  Google Scholar 

  59. Peggs KS, Quezada SA, Allison JP: Cell intrinsic mechanisms of T-cell inhibition and application to cancer therapy. Immunol Rev. 2008, 224: 141-165.

    CAS  PubMed  Google Scholar 

  60. Zitvogel L, Tesniere A, Kroemer G: Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006, 6: 715-727.

    CAS  PubMed  Google Scholar 

  61. Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Metivier D, Galluzzi L, Perfettini JL, Zitvogel L, Kroemer G: Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014, 21 (1): 79-91.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L: Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology. 2013, 2: e23510-

    PubMed  PubMed Central  Google Scholar 

  63. Maloney DG, Grillo-Lopez AJ, White CA, Bodkin D, Schilder RJ, Neidhart JA, Janakiraman N, Foon KA, Liles TM, Dallaire BK, Wey K, Royston I, Davis T, Levy R: IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma. Blood. 1997, 90: 2188-2195.

    CAS  PubMed  Google Scholar 

  64. Pegram MD, Konecny G, Slamon DJ: The molecular and cellular biology of HER2/neu gene amplification/overexpression and the clinical development of herceptin (trastuzumab) therapy for breast cancer. Cancer Treat Res. 2000, 103: 57-75.

    CAS  PubMed  Google Scholar 

  65. Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini JL, Castedo M, Mignot G, Panaretakis T, Casares N, Metivier D, Larochette N, van Endert P, Ciccosanti F, Piacentini M, Zitvogel L, Kroemer G: Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med. 2007, 13: 54-61.

    CAS  PubMed  Google Scholar 

  66. Shiao SL, Coussens LM: The tumor-immune microenvironment and response to radiation therapy. J Mammary Gland Biol Neoplasia. 2010, 15: 411-421.

    PubMed  PubMed Central  Google Scholar 

  67. Pardoll DM: Immunology beats cancer: a blueprint for successful translation. Nat Immunol. 2012, 13: 1129-1132.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Acharya UH, Jeter JM: Use of ipilimumab in the treatment of melanoma. Clin Pharmacol. 2013, 5: 21-27.

    PubMed  PubMed Central  Google Scholar 

  69. Devaud C, John LB, Westwood JA, Darcy PK, Kershaw MH: Immune modulation of the tumor microenvironment for enhancing cancer immunotherapy. Oncoimmunology. 2013, 2: e25961-

    PubMed  PubMed Central  Google Scholar 

  70. Parsa AT, Waldron JS, Panner A, Crane CA, Parney IF, Barry JJ, Cachola KE, Murray JC, Tihan T, Jensen MC, Mischel PS, Stokoe D, Pieper RO: Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007, 13: 84-88.

    CAS  PubMed  Google Scholar 

  71. Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, Takatsu Y, Melamed J, d'Adda di Fagagna F, Bernard D, Hernando E, Gil J: Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell. 2008, 133: 1006-1018.

    CAS  PubMed  Google Scholar 

  72. Beatty G, Paterson Y: IFN-gamma-dependent inhibition of tumor angiogenesis by tumor-infiltrating CD4+ T cells requires tumor responsiveness to IFN-gamma. J Immunol. 2001, 166: 2276-2282.

    CAS  PubMed  Google Scholar 

  73. Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS: Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell. 2008, 133: 1019-1031.

    CAS  PubMed  Google Scholar 

  74. Muller-Hermelink N, Braumuller H, Pichler B, Wieder T, Mailhammer R, Schaak K, Ghoreschi K, Yazdi A, Haubner R, Sander CA, Mocikat R, Schwaiger M, Forster I, Huss R, Weber WA, Kneilling M, Rocken M: TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell. 2008, 13: 507-518.

    PubMed  Google Scholar 

  75. Lawler PR, Lawler J: Molecular basis for the regulation of angiogenesis by thrombospondin-1 and -2. Cold Spring Harb Perspect Med. 2012, 2: a006627-

    PubMed  PubMed Central  Google Scholar 

  76. Li SS, Liu Z, Uzunel M, Sundqvist KG: Endogenous thrombospondin-1 is a cell-surface ligand for regulation of integrin-dependent T-lymphocyte adhesion. Blood. 2006, 108: 3112-3120.

    CAS  PubMed  Google Scholar 

  77. Baek KH, Bhang D, Zaslavsky A, Wang LC, Vachani A, Kim CF, Albelda SM, Evan GI, Ryeom S: Thrombospondin-1 mediates oncogenic Ras-induced senescence in premalignant lung tumors. J Clin Invest. 2013, 123: 4375-4389.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Sosale N, Discher DE: Marker-of-self becomes marker-of-senescence. Blood. 2012, 119: 5343-5344.

    CAS  PubMed  Google Scholar 

  79. Whitfield JR, Soucek L: Tumor microenvironment: becoming sick of Myc. Cell Mol Life Sci. 2012, 69: 931-934.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Galluzzi L, Vitale I, Kroemer G: Past, present, and future of molecular and cellular oncology. Front Oncol. 2011, 1: 1-

    PubMed  PubMed Central  Google Scholar 

  81. Somasundaram R, Villanueva J, Herlyn M: Intratumoral heterogeneity as a therapy resistance mechanism: role of melanoma subpopulations. Adv Pharmacol. 2012, 65: 335-359.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Al-Ejeh F, Smart CE, Morrison BJ, Chenevix-Trench G, Lopez JA, Lakhani SR, Brown MP, Khanna KK: Breast cancer stem cells: treatment resistance and therapeutic opportunities. Carcinogenesis. 2011, 32: 650-658.

    CAS  PubMed  Google Scholar 

  83. Soucek L, Lawlor ER, Soto D, Shchors K, Swigart LB, Evan GI: Mast cells are required for angiogenesis and macroscopic expansion of Myc-induced pancreatic islet tumors. Nat Med. 2007, 13: 1211-1218.

    CAS  PubMed  Google Scholar 

  84. Reimann M, Lee S, Loddenkemper C, Dorr JR, Tabor V, Aichele P, Stein H, Dorken B, Jenuweins T, Schmitt CA: Tumor Stroma-Derived TGF-beta Limits Myc-Driven Lymphomagenesis via Suv39h1-Dependent Senescence. Cancer Cell. 2010, 17: 262-272.

    CAS  PubMed  Google Scholar 

  85. Prestwich RJ, Errington F, Hatfield P, Merrick AE, Ilett EJ, Selby PJ, Melcher AA: The immune system–is it relevant to cancer development, progression and treatment?. Clin Oncol (R Coll Radiol). 2008, 20: 101-112.

    CAS  Google Scholar 

  86. Hannani D, Sistigu A, Kepp O, Galluzzi L, Kroemer G, Zitvogel L: Prerequisites for the antitumor vaccine-like effect of chemotherapy and radiotherapy. Cancer J. 2011, 17: 351-358.

    CAS  PubMed  Google Scholar 

  87. Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, Tesniere A, Martins I, Ly A, Haynes NM, Smyth MJ, Kroemer G, Zitvogel L: Chemotherapy and radiotherapy: cryptic anticancer vaccines. Semin Immunol. 2010, 22: 113-124.

    PubMed  Google Scholar 

  88. Dougan M, Dranoff G: Immune therapy for cancer. Annu Rev Immunol. 2009, 27: 83-117.

    CAS  PubMed  Google Scholar 

  89. Menard C, Blay JY, Borg C, Michiels S, Ghiringhelli F, Robert C, Nonn C, Chaput N, Taieb J, Delahaye NF, Flament C, Emile JF, Le Cesne A, Zitvogel L: Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 2009, 69: 3563-3569.

    CAS  PubMed  Google Scholar 

  90. Wilmott JS, Long GV, Howle JR, Haydu LE, Sharma RN, Thompson JF, Kefford RF, Hersey P, Scolyer RA: Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma. Clin Cancer Res. 2012, 18: 1386-1394.

    CAS  PubMed  Google Scholar 

  91. Wilmott JS, Scolyer RA, Long GV, Hersey P: Combined targeted therapy and immunotherapy in the treatment of advanced melanoma. Oncoimmunology. 2012, 1: 997-999.

    PubMed  PubMed Central  Google Scholar 

  92. Liu C, Peng W, Xu C, Lou Y, Zhang M, Wargo JA, Chen J, Li HS, Watowich S, Yang Y, Frederick DT, Cooper ZA, Mbofung R, Whittington M, Flaherty KT, Woodman SE, Davies MA, Radvanyi LG, Overwijk WW, Lizee G, Hwu P: BRAF Inhibition Increases Tumor Infiltration by T cells and Enhances the Anti-tumor Activity of Adoptive Immunotherapy in Mice. Clin Cancer Res. 2013, 19 (2): 393-403.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Sumimoto H, Imabayashi F, Iwata T, Kawakami Y: The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J Exp Med. 2006, 203: 1651-1656.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Bajor DL, Vonderheide RH: Rehabilitation for oncogene addiction: role of immunity in cellular sobriety. Clin Cancer Res. 2012, 18: 1192-1194.

    PubMed  Google Scholar 

  95. Knight DA, Ngiow SF, Li M, Parmenter T, Mok S, Cass A, Haynes NM, Kinross K, Yagita H, Koya RC, Graeber TG, Ribas A, McArthur GA, Smyth MJ: Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J Clin Invest. 2013, 123: 1371-1381.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Bex A, Etto T, Vyth-Dreese F, Blank C, Griffioen AW: Immunological heterogeneity of the RCC microenvironment: do targeted therapies influence immune response?. Curr Oncol Rep. 2012, 14: 230-239.

    CAS  PubMed  Google Scholar 

  97. Finke JH, Rini B, Ireland J, Rayman P, Richmond A, Golshayan A, Wood L, Elson P, Garcia J, Dreicer R, Bukowski R: Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients. Clin Cancer Res. 2008, 14: 6674-6682.

    CAS  PubMed  Google Scholar 

  98. Busse A, Asemissen AM, Nonnenmacher A, Braun F, Ochsenreither S, Stather D, Fusi A, Schmittel A, Miller K, Thiel E, Keilholz U: Immunomodulatory effects of sorafenib on peripheral immune effector cells in metastatic renal cell carcinoma. Eur J Cancer. 2011, 47: 690-696.

    CAS  PubMed  Google Scholar 

  99. Zhang H, Melamed J, Wei P, Cox K, Frankel W, Bahnson RR, Robinson N, Pyka R, Liu Y, Zheng P: Concordant down-regulation of proto-oncogene PML and major histocompatibility antigen HLA class I expression in high-grade prostate cancer. Cancer Immun. 2003, 3: 2-

    PubMed  Google Scholar 

  100. Chang CL, Hsu YT, Wu CC, Yang YC, Wang C, Wu TC, Hung CF: Immune mechanism of the antitumor effects generated by bortezomib. J Immunol. 2012, 189: 3209-3220.

    CAS  PubMed  Google Scholar 

  101. Guttman-Yassky E, Mita A, De Jonge M, Matthews L, McCarthy S, Iwata KK, Verweij J, Rowinsky EK, Krueger JG: Characterisation of the cutaneous pathology in non-small cell lung cancer (NSCLC) patients treated with the EGFR tyrosine kinase inhibitor erlotinib. Eur J Cancer. 2010, 46: 2010-2019.

    CAS  PubMed  Google Scholar 

  102. Jaime-Ramirez AC, Mundy-Bosse BL, Kondadasula S, Jones NB, Roda JM, Mani A, Parihar R, Karpa V, Papenfuss TL, LaPerle KM, Biller E, Lehman A, Chaudhury AR, Jarjoura D, Burry RW, Carson WE: IL-12 enhances the antitumor actions of trastuzumab via NK cell IFN-gamma production. J Immunol. 2011, 186: 3401-3409.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski D, Colevas AD, Weng WK, Clarke MF, Carlson RW, Stockdale FE, Mollick JA, Chen L, Levy R: Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer. J Clin Invest. 2012, 122: 1066-1075.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Boni A, Cogdill AP, Dang P, Udayakumar D, Njauw CN, Sloss CM, Ferrone CR, Flaherty KT, Lawrence DP, Fisher DE, Tsao H, Wargo JA: Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function. Cancer Res. 2010, 70: 5213-5219.

    CAS  PubMed  Google Scholar 

  105. Catellani S, Pierri I, Gobbi M, Poggi A, Zocchi MR: Imatinib treatment induces CD5+ B lymphocytes and IgM natural antibodies with anti-leukemic reactivity in patients with chronic myelogenous leukemia. PLoS One. 2011, 6: e18925-

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Krusch M, Salih HR: Effects of BCR-ABL inhibitors on anti-tumor immunity. Curr Med Chem. 2011, 18: 5174-5184.

    CAS  PubMed  Google Scholar 

  107. Ohyashiki K, Katagiri S, Tauchi T, Ohyashiki JH, Maeda Y, Matsumura I, Kyo T: Increased natural killer cells and decreased CD3(+)CD8(+)CD62L(+) T cells in CML patients who sustained complete molecular remission after discontinuation of imatinib. Br J Haematol. 2012, 157: 254-256.

    CAS  PubMed  Google Scholar 

  108. Kreutzman A, Juvonen V, Kairisto V, Ekblom M, Stenke L, Seggewiss R, Porkka K, Mustjoki S: Mono/oligoclonal T and NK cells are common in chronic myeloid leukemia patients at diagnosis and expand during dasatinib therapy. Blood. 2010, 116: 772-782.

    CAS  PubMed  Google Scholar 

  109. Chen J, Schmitt A, Giannopoulos K, Chen B, Rojewski M, Dohner H, Bunjes D, Schmitt M: Imatinib impairs the proliferation and function of CD4+CD25+ regulatory T cells in a dose-dependent manner. Int J Oncol. 2007, 31: 1133-1139.

    CAS  PubMed  Google Scholar 

  110. Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, Yu Z, Rosenberg SA, Restifo NP: Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother. 2010, 33: 1-7.

    PubMed  PubMed Central  Google Scholar 

  111. Humphrey RW, Brockway-Lunardi LM, Bonk DT, Dohoney KM, Doroshow JH, Meech SJ, Ratain MJ, Topalian SL, Pardoll DM: Opportunities and challenges in the development of experimental drug combinations for cancer. J Natl Cancer Inst. 2011, 103: 1222-1226.

    CAS  PubMed  Google Scholar 

  112. Vanneman M, Dranoff G: Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer. 2012, 12: 237-251.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Mellman I, Coukos G, Dranoff G: Cancer immunotherapy comes of age. Nature. 2011, 480: 480-489.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. GlaxoSmithKline: Study of Dabrafenib +/- Trametinib in Combination With Ipilimumab for V600E/K Mutation Positive Metastatic or Unresectable Melanoma. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01767454 NLM Identifier: NCT01767454

    Google Scholar 

  115. National Cancer Institute: Ipilimumab With or Without Dabrafenib, and/or Trametinib in Treating Patients With Melanoma That is Metastatic or Cannot Be Removed By Surgery. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01940809 NLM Identifier: NCT01940809

    Google Scholar 

  116. Tarhini AA, Frankel P, Margolin KA, Christensen S, Ruel C, Shipe-Spotloe J, Gandara DR, Chen A, Kirkwood JM: Aflibercept (VEGF Trap) in inoperable stage III or stage iv melanoma of cutaneous or uveal origin. Clin Cancer Res. 2011, 17: 6574-6581.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Azijli K, Stelloo E, Peters GJ, AJ VDE: New developments in the treatment of metastatic melanoma: immune checkpoint inhibitors and targeted therapies. Anticancer Res. 2014, 34: 1493-1505.

    CAS  PubMed  Google Scholar 

  118. National Cancer Institute: Ipilimumab and Brentuximab Vedotin in Treating Patients With Relapsed or Refractory Hodgkin Lymphoma. ClinicalTrials.gov Identifier: NCT01896999. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01896999 NLM Identifier: NCT01896999

    Google Scholar 

  119. University of Utah: Ipilimumab Plus Targeted Inhibitor (Erlotinib or Crizotinib) for EGFR or ALK Mutated Stage IV Non-small Cell Lung Cancer: Phase Ib With Expansion Cohorts. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01998126 NLM Identifier: NCT01998126

    Google Scholar 

  120. Hoffman-LaRoche: A Phase 1b Study of MPDL3280A (an Engineered Anti-PDL1 Antibody) in Combination With Tarceva in Patients With Non-Small Cell Lung Cancer. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT02013219 NLM Identifier: NCT02013219

    Google Scholar 

  121. Richardson PG, Weller E, Lonial S, Jakubowiak AJ, Jagannath S, Raje NS, Avigan DE, Xie W, Ghobrial IM, Schlossman RL, Mazumder A, Munshi NC, Vesole DH, Joyce R, Kaufman JL, Doss D, Warren DL, Lunde LE, Kaster S, Delaney C, Hideshima T, Mitsiades CS, Knight R, Esseltine DL, Anderson KC: Lenalidomide, bortezomib, and dexamethasone combination therapy in patients with newly diagnosed multiple myeloma. Blood. 2010, 116: 679-686.

    CAS  PubMed  PubMed Central  Google Scholar 

  122. National Cancer Institute: Lenalidomide and Ibrutinib in Treating Patients With Relapsed or Refractory Chronic Lymphocytic Leukemia or Small Lymphocytic Lymphoma. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01886859 NLM Identifier: NCT01886859

    Google Scholar 

  123. Bristol-Meyers Squibb: Nivolumab (BMS-936558; MDX-1106) in Combination With Sunitinib, Pazopanib, or Ipilimumab in Subjects With Metastatic Renal Cell Carcinoma (RCC) (CheckMate 016). ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01472081 NLM Identifier: NCT01472081

    Google Scholar 

  124. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O'Toole T, Lustgarten S, Moore L, Motzer RJ: Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007, 356: 2271-2281.

    CAS  PubMed  Google Scholar 

  125. Guilhot F, Roy L, Martineua G, Guilhot J, Millot F: Immunotherapy in chronic myelogenous leukemia. Clin Lymphoma Myeloma. 2007, 7 (Suppl 2): S64-70.

    CAS  PubMed  Google Scholar 

  126. Westin JR, Chu F, Zhang M, Fayad LE, Kwak LW, Fowler N, Romaguera J, Hagemeister F, Fanale M, Samaniego F, Feng L, Baladandayuthapani V, Wang Z, Ma W, Gao Y, Wallace M, Vence LM, Radvanyi L, Muzzafar T, Rotem-Yehudar R, Davis RE, Neelapu SS: Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol. 2014, 15: 69-77.

    CAS  PubMed  PubMed Central  Google Scholar 

  127. San Antonio Military Medical Center: Combination Immunotherapy With Herceptin and the HER2 Vaccine NeuVax. ClinicalTrials.gov [Internet]. 2000–, Bethesda (MD): National Library of Medicine (US), [cited 2014 May 21] Available from: http://clinicaltrials.gov/show/NCT01570036 NLM Identifier: NCT01570036

    Google Scholar 

Download references

Acknowledgements

The authors acknowledge current and former members of the Felsher laboratory. Within the Felsher laboratory, research has been funded by the Burroughs Welcome Fund Career Award, the Damon Runyon Foundation Lilly Clinical Investigator Award, NIH NCI K23 CA140722 to A.C.F., NIH RO1 grant number CA 089305, 105102, 170378 PQ22, U54CA149145, U54CA143907, National Cancer Institute’s In-vivo Cellular and Molecular Imaging Center grant number CA 114747, Integrative Cancer Biology Program grant number CA 112973, NIH/NCI PO1 grant number CA034233, and the Leukemia and Lymphoma Society Translational Research grant number R6223-07. S.C.C. was previously supported by NIH, 5 T32 AI07290 and is currently supported by an NIH NRSA from the NCI (F32CA177139).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dean W Felsher.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SCC and DWF conceived of and wrote the review. YL helped write the review. ACF provided a clinical perspective on targeted and immunological therapies. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Casey, S.C., Li, Y., Fan, A.C. et al. Oncogene withdrawal engages the immune system to induce sustained cancer regression. j. immunotherapy cancer 2, 24 (2014). https://doi.org/10.1186/2051-1426-2-24

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/2051-1426-2-24

Keywords