Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Nov 1.
Published in final edited form as: Trends Cancer. 2023 Jul 29;9(11):928–936. doi: 10.1016/j.trecan.2023.07.004

Immune vulnerabilities of mutant KRAS in pancreatic cancer

Noah C Cheng 1, Robert H Vonderheide 1,*
PMCID: PMC10592263  NIHMSID: NIHMS1921440  PMID: 37524642

Abstract

The 40-year desire to target mutant KRAS (mKRAS) therapeutically is being realized with more and more broadly applicable and tumor-specific small molecular inhibitors. Immunologically, mKRAS has equal desirability as a target. Tumor KRAS signaling plays a large role in shaping the immunosuppressive nature of the tumor microenvironment, especially in pancreatic cancer, leaving mKRAS inhibitors with potentially powerful immune modulatory capabilities that could be exploited in immune-oncology combinations. mKRAS is itself an immunological antigen, a “shared neo-epitope” linked to the oncogenic process, validated biochemically and immunologically. Novel approaches in the clinic are taking advantage of the fact that mKRAS peptides are naturally processed and presented in tumors by MHC.

Keywords: Pancreatic cancer, mutant KRAS, inhibitors, vaccines, TCR therapies, immune response

Pancreatic cancer and its driving oncogene mutant KRAS

Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second leading cause of US cancer-related deaths by 2030 [1], and despite therapeutic progress, the 5-year overall survival rate for PDAC is only 12% [2] largely due to insufficient early detection and poorly effective treatment strategies of advanced disease. First-line treatment for patients with metastatic pancreatic cancer remains combination chemotherapy, which has relatively short durability and high toxicity [3, 4]. Despite successes in patients with other cancers, immunotherapies, such as immune checkpoint antibodies, have minimal if any efficacy in PDAC as single-agents [5, 6] and mixed results when combined with chemotherapy [7].

Once thought to be undruggable, mutant Kirsten Ras (mKRAS) has become an increasingly tractable therapeutic target in patients with pancreatic cancer [8]. Oncogenic, activating KRAS mutations are present in >90% of PDAC cases [9]. KRAS is a GTPase that regulates proliferation and survival, among other cell functions, and is rendered constitutively active by genetic point mutations [10]. mKRAS is an ideal drug target in PDAC for several reasons. First, KRAS mutations initiate the formation of pre-cancerous lesions of the pancreas, termed pancreatic intraepithelial neoplasia (PanIN), and drive the development and growth of invasive carcinomas. Second, mutations in the KRAS gene are specific to tumor cells, limiting the potential off-target activity of any anti-mKRAS therapy. Lastly, the high prevalence of KRAS mutations in PDAC gives rise to a large patient population that can benefit from mKRAS treatments. In this review, we discuss how work from our group and many others has demonstrated that mKRAS shapes the tumor immune microenvironment (TME), can be targeted with small molecule inhibitors that engender immunological effects, and is a neoantigen in PDAC.

The KRAS oncogene orchestrates the pancreatic immune microenvironment

Mouse models of PDAC suggest that tumors arise in an “immune privileged” niche in which immunosuppression is a primary, rather than secondary, event. In the classic immunoediting theory, tumor cells are initially eliminated until immunosuppressive pathways are activated [11]. However, dysfunctional dendritic cells, suboptimal T cell priming, and high levels of immunosuppression in PanINs suggest that PDAC diverges from the immunoediting theory because of its immunologically “cold” status even at the earliest stages of tumorigenesis [1216].

mKRAS, also referred to as oncogenic KRAS here, promotes the accumulation of immunosuppressive cells that exclude cytotoxic T cells (Figure 1), as demonstrated in autochthonous genetically engineered mouse models of PDAC that recapitulate the histopathological and clinical features of human PDAC. These models include the KrasLSL-G12D/+ Pdx1-Cre (KC) and KrasLSL-G12D/+ Trp53LSL-R172H/+ Pdx1-Cre (KPC) mice [17, 18]. Using KC mice, we demonstrated that PanIN formation is accompanied by an influx of immunosuppressive cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). This infiltration of immunosuppressive cells is accompanied by a dearth of activated, effector T cells [19]. Oncogenic KRAS upregulates tumor cytokine expression, such as GM-CSF, which leads to the accumulation of MDSCs [20, 21]. These MDSCs actively participate in the exclusion of CD8+ T cells from the PanIN and tumor microenvironments, because their depletion or the neutralization of GM-CSF enhances T cell infiltration. In an inducible mKRAS model with pancreatitis, KRAS expression causes PanIN infiltration of CD4+ T cells (most likely Tregs), which can also regulate inflammatory cell infiltration and inhibit CD8+ T cell activation [22]. mKRAS signaling plays a critical role in recruiting immunosuppressive cells and potentially even converting cells to such a phenotype [23].

Figure 1. Mutant (m)KRAS establishes an immunosuppressive microenvironment in the pancreas throughout tumorigenesis.

Figure 1.

Expression of mKRAS leads to the recruitment of immunosuppressive cells, such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), that inhibit CD8+ T cell infiltration and function. GM-CSF release is upregulated by mKRAS and directly modulates MDSC accumulation. mKRAS signaling in pre-cancerous, neoplastic epithelial cells promotes cancer-associated fibroblast (CAF) accumulation and expression of cytokines (CXCL1, Il-6, IL-33, and SAA3) that support immunosuppressive myeloid cell populations. mKRAS also primes pancreatic neoplastic and tumor cells to respond to pro-tumorigenic cytokines released by inflammatory immune cells. mKRAS induces expression of IL-17R and type I cytokine receptors, IL-4Rα and IL-13Rα1, enabling the neoplastic/tumor cells to respond to growthpromoting cytokines released by Th17 and Th2 cells, respectively. Increased IL-6 production following KRAS mutation helps maintain a pool of Th17 cells in the pancreatic microenvironment. This figure was made using Biorender.com. Abbreviations: GM-CSF, granulocyte-macrophage colony-stimulating factor; CXCL1, C-X-C motif chemokine ligand 1; IL-6, interleukin-6; IL-33, interleukin-33; SAA3, serum amyloid a3; IL-17(R), interleukin-17 (receptor); IL-4(Rα), interleukin-4 (receptor α); IL-13(Rα1) interleukin-13 (receptor α 1); Th17, T helper 17; Th2, T helper 2.

The desmoplasia characteristic of PDAC includes cancer-associated fibroblasts (CAFs), which are immune-modulating and influenced by mKRAS signaling [24]. In the inducible mouse model of KRASG12D, oncogenic KRAS promotes infiltration, activation, and reprogramming of CAFs at PanIN stages (Figure 1). Specifically, mKRAS signaling in transforming epithelial cells extrinsically programs CAFs to produce cytokines (i.e. IL-33 and IL-6) that regulate immunosuppressive myeloid cells [25]. In established PDAC tumors, CAFs, depending on the subtype, can be immune-suppressive (antigen-presenting and LRRC15+ CAFs) [26, 27] or immune-enhancing (T CAFs) [28]. The exact immune-mediating function of CAFs in PDAC is likely also based on the stage of carcinogenesis.

T helper 17 (Th17) cells have a clearly defined role in inflammation and autoimmunity, but their involvement in cancer and tumor immunity is more nuanced [29]. Oncogenic KRAS expression drives the production of IL-6 [30, 31], a polarizing cytokine for Th17 cells, and helps orchestrate the infiltration of Th17 cells [32]. mKRAS signaling also upregulates IL-17R on PanIN epithelial cells in a cell-autonomous manner, and Th17 cells release IL-17 that promotes PanIN formation and progression [32]. A similarly constructed pathway exists involving Th2 cells. In an inducible KRASG12D PDAC model, mKRAS increases expression of type I cytokine receptors, such as IL-4Rα and IL-13Rα1, which recognize IL-4 and IL-13 secreted by PanIN and tumor infiltrating Th2 cells. These cytokines are tumor-promoting as they upregulate MYC through the JAK1-STAT6 pathway [33]. In both of these examples, downstream effects of mutant KRAS expression give neoplastic epithelial or tumor cells the ability to respond to pro-tumorigenic signals coming from the inflammatory immune cell milieu (Figure 1).

Studies that inactivate KRAS reaffirm the immune modulatory role of the oncogene and tantalizingly suggest that targeting mKRAS therapeutically might synergize with immunotherapy approaches in PDAC. In the inducible KRASG12D model, turning KRASG12D off repolarizes macrophages away from an immunosuppressive M2 phenotype and decreases the number of pancreatic-infiltrating MDSCs [25, 34]. In a CRISPR/Cas9 experimental system that knock-outs (KO) KRASG12D in a PDAC cell line [35], KRAS KO orthotopic tumors exhibit polarization of TAMs to an anti-tumor M1 state and increased infiltration of CD4+ and cytotoxic CD8+ T cells. These T cells are more active and produce IFNγ. Among other effects in the TME, IFNγ induces tumor cell apoptosis, angiogenesis inhibition, and activation of M1 macrophages, which are capable of specifically killing tumor cells [3638]. Interestingly, these KRAS KO tumor cells have increased expression of major histocompatibility complex (MHC), consistent with improved immunogenicity [35]. Evidently, targeting mKRAS can reverse the immunosuppression inherent to PDAC and possibly augment immunotherapies.

Although knocking out mKRAS in PDAC tumors in vivo has been attempted [39], the predominant approach to blocking oncogenic KRAS expression in a therapeutic setting is through small molecule inhibitors (Table 1). The recent identification and characterization of KRASG12C inhibitors have shown that drugging mKRAS is possible [4043]. These KRASG12C inhibitors exhibit therapeutic efficacy in various solid tumors, including pancreatic cancer [4447]; however, in PDAC, KRASG12C mutations are present in only 1% of patients, whereas KRASG12D represents ~40% of all KRAS mutations [48]. We recently investigated the efficacy of a novel, non-covalent inhibitor of KRASG12D (MRTX1133) in immunocompetent mouse models of PDAC [49], as the initial studies interrogated the drug in immune deficient hosts [50, 51].

Table 1.

Advantages and drawbacks of mKRAS targeting therapeutics in pancreatic cancera

Anti-mKRAS therapy Advantages Drawbacks
Small molecule inhibitors
  • Potent anti-tumor activity in models and patients

  • ↑ MHC expression

  • ↓ Immunosuppressive cells

  • ↑ Effector T cell infiltration and functionality

  • Potential synergy with immunotherapies

  • Development of resistance

  • Potential impact on normal cells

Vaccines
  • Pooled peptide/mRNA vaccines can be made to cover several variants

  • Can induce long-lasting anti-tumor immunity

  • Must overcome immunosuppression in the TME

ACT with engineered TCRs
  • Promising early clinical case reports

  • Can induce long-lasting anti-tumor immunity

  • Must overcome immunosuppression in the TME

  • MHC restricted

  • Dependent on sufficient antigen density and MHC expression

  • Highly personalized so costly and laborious to produce

Bispecific antibodies
  • Can have very high affinity for mKRAS without compromising specificity

  • Personalized yet off-the-shelf

  • MHC restricted

  • Dependent on sufficient antigen density and MHC expression

Hapimmune
  • Combines benefits of inhibitor and TCR-based therapies: direct inhibition, favorable immunomodulation, and immune-mediated killing

  • Dependent on sufficient antigen density and MHC expression

a

Abbreviations: ACT, adoptive T cell therapy; HLA, human leukocyte antigen; MHC, major histocompatibility complex; mKRAS, mutant KRAS; TCR, T cell receptor; TME, tumor microenvironment.

MRTX1133 shows robust efficacy in subcutaneous, orthotopic, and autochthonous models. In fact, we have never observed such pronounced and lasting tumor regressions in KPC mice in our many years of testing anti-tumor agents in this model. Interestingly, the drug’s full efficacy is dependent on T cells, and inhibiting mKRAS favorably alters the PDAC tumor immune microenvironment and improves anti-tumor immunity. MRTX1133 leads to increased expression of MHC class I on tumor cells, increased αSMA+ fibroblasts, repolarization of macrophages to an M1 phenotype, decreased proportion of MDSCs, and increased infiltration of T cells. Moreover, these T cells are more proliferative, activated, and cytotoxic [49]. Our results suggest that mKRAS inhibitors could not only reverse the immunosuppression constructed by oncogenic KRAS expression but also induce a “vaccine effect” by which T cells are primed to restrain future tumor regrowth. In all, KRASG12D inhibitors have immense promise to treat a large proportion of PDAC patients and open the door for new combinatorial treatment strategies.

Preclinical studies suggest synergy between KRASG12C inhibitors and anti-PD-1 therapy, although none was performed in the context of PDAC [43, 52, 53]. Clinical trials are underway to test KRASG12C inhibitors in combination with immune checkpoint blockade in non-small cell lung cancer patients (NCT04185883 and NCT04613596), and early results indicate manageable adverse events and encouraging efficacy [54]. Beyond immune checkpoint inhibitors, other immune agents that may improve T cell priming, such as Flt3L and agonistic CD40 [13, 55], are potential combination partners in PDAC, as are vaccines and adoptive T cell therapies, both of which will be discussed later in this article. One possible combinatorial strategy involves the “hapimmune” concept (Table 1). The cysteine made available by the KRASG12C mutation enables the production of covalent inhibitors. These inhibitors bind mKRAS; mKRAS-inhibitor complexes are processed and presented in the context of MHC; then, the drug-peptide conjugates can be targeted with bispecific antibodies and potentially T cell receptors (TCRs) [56, 57]. The development of covalent (and non-covalent) inhibitors for other mKRAS variants, especially KRASG12D, would encourage further testing of the hapimmune approach with more relevance to PDAC.

Vaccination against mutant KRAS

The specificity of oncogenic KRAS to PDAC cells and its importance in tumor growth makes the mutated protein an attractive tumor neoantigen that can be targeted by T cells. The discovery of naturally occurring mKRAS-specific T cells opened the possibility for vaccination and adoptive T cell therapy strategies. Some of the earliest studies observed KRASG12V-specific CD4+ T cells in healthy individuals [58] and KRASG13D-specific CD4+ and CD8+ T cells in a colorectal cancer patient [59]. Another analysis found that a natural reservoir of mKRAS-specific CD4+ and CD8+ T cells exists in human leukocyte antigen (HLA)-A*02-positive PDAC patients [60]. Following stimulation, these CD8+ T cells are able to kill KRASG12V-mutated pancreatic cancer cells in an HLA-restricted manner. Identification of naturally occurring anti-mKRAS T cell responses prompted a series of mKRAS vaccine trials.

mKRAS vaccines have been shown to induce mKRAS-specific immunity but their efficacy and the best setting for use remains uncertain (Table 1). A pioneering trial vaccinated a small cohort of pancreatic cancer patients with peripheral blood mononuclear cells (PBMCs) loaded with synthetic mKRAS peptides [61]. Although no therapeutic benefit was observed, oncogenic KRAS-specific T cells were induced by vaccination in a couple of patients, and in one of the patients, these T cells were able to kill autologous tumor cells in an HLA-restricted manner in vitro [62]. In another study using PBMCs pulsed with mKRAS peptides, only one out of nine pancreatic cancer patients responded to the vaccine [63]. mKRAS peptide vaccines have also been tested with adjuvants, such as GM-CSF. A considerable number of patients with advanced PDAC who received synthetic mKRAS peptide(s) and GM-CSF demonstrated an immunological response [64]. Moreover, a subset of these immune responders had stable disease following treatment, and responders exhibited a survival benefit when compared to those that did not respond to vaccination. In the adjuvant setting, peptide-based vaccination against mKRAS has shown more clinical promise but the therapeutic efficacy has still not been confirmed [6567]. Interestingly, one study found memory T cell responses in patients up to 9 years following mKRAS peptide vaccine adjuvant therapy [67]. Recently, a phase I clinical trial (NCT03948763) was initiated to test an anti-mKRAS mRNA-based vaccine (mRNA-5671/V941) with or without anti-PD-1 therapy in patients with advanced or metastatic PDAC, non-small cell lung cancer, or colorectal cancer. Enrollment criteria include a confirmed KRAS mutation (G12D, G12V, G13D, or G12C) and HLA-A*11:01 and/or HLA-C*08:02 positivity. Another trial (NCT03953235) is investigating an adenovirus and self-amplifying mRNA mKRAS vaccine regimen in combination with immune checkpoint blockade (ICB) in PDAC and other solid tumors. Future mKRAS vaccine studies in PDAC should consider combination approaches with immunotherapies other than ICB, which exhibits limited efficacy in the disease.

Given that KRAS mutations are usually the initial and driving mutation in PanIN formation and PDAC development, vaccinating high-risk – but healthy – individuals offers a promising approach to preventing, or intercepting, PDAC. Attenuated Listeria monocytogenes (LM) engineered to express KRASG12D in combination with a Treg-depleting regimen prolongs survival and delays PanIN development when given to KPC mice bearing early-stage, but not late-stage, PanINs [68]. These data provide evidence that intervening in the natural history of PDAC using a mKRAS-based therapy is feasible. A current phase 1 clinical trial (NCT05013216) is testing a pool of mKRAS peptides as a vaccine for individuals who are at high risk of developing PDAC but who have not done so yet. Enrollment criteria include a family history of pancreatic cancer and/or predisposing germline mutations as well as imaging of a pancreatic abnormality, such as a cyst. While a potential therapeutic benefit for such an approach will not reveal itself for many years, determining whether mKRAS-targeting T cell responses can be generated at precancerous, neoplastic stages is important.

It seems logical that mKRAS vaccination will have to be used and tested in combination with other therapies, beyond immune checkpoint blockade, that manipulate the immunosuppression inherent to PDAC. Oncogenic KRAS constructs a pancreatic microenvironment that inhibits a natural anti-tumor or anti-PanIN immune response. Thus, even if a vaccine could stimulate the expansion of mKRAS-specific T cells, there is no guarantee that those T cells could overcome the immunosuppressive barriers set in place in vivo. Indeed, for a vaccine to have efficacy in PanIN-bearing KPC mice, a Treg-depleting treatment that is not readily translatable is needed [68]. Perhaps these vaccines will prove most efficacious in the adjuvant setting when tumor burden is low [69]; otherwise, a focus could be put on interrogating mKRAS vaccines in combination with mKRAS inhibitors and/or immunotherapies proven to remodel the PDAC tumor immune microenvironment.

Targeting mutant KRAS using TCR-based therapies

A growing body of work has identified HLA class I-restricted mKRAS epitopes and TCRs that recognize these epitopes. T cells engineered to express these TCRs show therapeutic efficacy in preclinical mouse models. For example, mKRAS-specific TCRs have been isolated from HLA-A*11:01 transgenic mice immunized with KRASG12D/V peptides [70]. Human T cells transduced with the KRASG12D-reactive TCR slow tumor growth and prolong survival in immunodeficient mice implanted with a human HLA-A*11:01- and KRASG12D-positive pancreatic cancer cell line. We and our colleagues used a bioinformatic, biochemical, and proteomic approach to characterize mKRAS G12 epitopes that are processed and then presented with high affinity and stability by three HLA class I complexes: HLA-A*03:01, HLA-A*11:01, and HLA-B*07:02 [71]. TCRα/β pairs were then isolated and sequenced from CD8+ T cells that recognized immunogenic mKRAS epitopes. CD8+ T cells transduced with TCRs specific for KRASG12V in the context of HLA-A*03:01 or HLA-A*11:01 exhibit lytic activity against HLA class I matched human tumor cell lines and have potent anti-tumor activity when adoptively transferred into immunodeficient mice harboring metastatic lung cancer. A similar pipeline was taken (1) to identify even more mKRAS neoantigens (4 KRAS mutations across 9 common HLA class I alleles) that are endogenously processed and presented and (2) to broaden the repertoire of mKRAS peptide-HLA class I complexes that are known to be immunogenic [72]. mKRAS-reactive TCRs can also be screened from tumor-infiltrating and peripheral blood lymphocytes of cancer patients with KRAS-mutated tumors [73]. Once identified, mKRAS TCRs can form the basis of adoptive T cell therapy and also bispecific, T cell engaging molecules that fuse TCRs or TCR-mimic single-chain variable fragments (scFvs) to an anti-CD3 scFv, as has been previously shown [74, 75] (Table 1).

Initial clinical observations have demonstrated the promise of TCR T cell therapies targeting mKRAS. In one case, a colon cancer patient showed a natural T cell response to KRASG12D [76]. Upon infusion of autologous T cells engineered to express an HLA-C*08:02-restricted TCR specific for KRASG12D, the patient had a 9-month partial response and regressions of lung metastases. In a subsequent case, a patient with metastatic pancreatic cancer was given autologous T cells transduced with KRASG12D-specific, HLA-C*08:02-restricted TCRs [77]. The lung metastases of this patient regressed. These studies serve as proof-of-concept for the therapeutic potential for mKRAS TCR therapy and provide promise for larger clinical trials (NCT03190941 and NCT03745326).

To bring mKRAS T cell therapies to a larger patient population, identification of more mKRAS epitope and HLA class I peptidomes as well as TCRs that recognize such complexes will be critical. While it has been shown that mKRAS neoantigens are presented by some of the most common HLA class I alleles, such as HLA-A*03:01 and HLA-A*11:01 [7072], there are many other HLA class I molecules that, to our knowledge, have not been thoroughly investigated in terms of mKRAS epitopes. Established pipelines for mKRAS neoantigen discovery can be used to comprehensively interrogate both HLA class I and II alleles of all frequencies to find which ones naturally present mKRAS epitopes and which peptide-HLA complexes are immunogenic [78].

Even as more candidate mKRAS neoantigens are found, the low abundance of these neoantigens presents a challenge for TCR-based therapies. For example, it has been estimated that the number of KRASG12V peptides presented on various cancer cell lines is no greater than ~100 per cell and can be as low as ~10 per cell [71, 72, 75]. The activity of TCR-engineered cells correlates with mKRAS peptide-HLA complex abundance [71], so elucidating ways to increase activity will be important. Increasing the avidity of mKRAS TCRs is one such strategy. In fact, the affinity of a KRASG12D-specific TCR was enhanced one million-fold without losing any specificity for the mutated variant over the wild-type one [74].

TCR-based therapies, of course, are dependent on peptide-MHC expression, which can be problematic. For the patient with metastatic colon cancer noted above who was given mKRAS-specific T cells, the one lung lesion that progressed lost expression of the MHC molecule presenting KRASG12D [76]. Pancreatic cancer has established mechanisms that decrease antigen presentation, such as the downregulation of MHC class I expression [79, 80]. Autophagy has been shown to selectively degrade MHC class I molecules in PDAC and thus likely contributes to the deficient antigen presentation in this malignancy [81]. Inhibiting autophagy rescues MHC class I expression and enhances the efficacy of immune checkpoint blockade in a mouse model of PDAC. Combining autophagy inhibitors with mKRAS TCR-engineered T cells is an intriguing therapeutic avenue, as the autophagy inhibitors could increase the number of targets for T cell therapy.

Concluding remarks

mKRAS has long been an attractive therapeutic target in pancreatic cancer given its ubiquity in the patient population, its specificity to the tumor, and its important role in tumor development. Recent work has shown that (1) mKRAS is druggable with small molecule inhibitors and (2) the oncogenic protein is an immunological target itself. mKRAS inhibitors may have immunologic consequences, reversing mKRAS-constructed immunosuppression in the pancreatic microenvironment and cooperating with T cell immunity for full effect. Complimentarily, vaccines and T cell receptor-based therapies are being developed to induce anti-mKRAS immune responses. Nonetheless, many knowledge gaps and questions remain (see Outstanding questions), such as the clinical efficacy of and proper setting for mKRAS vaccines. Future work must focus on identifying potent and viable treatment combinations (i.e. mKRAS inhibitors with immune checkpoint blockade or mKRAS inhibitors with TCR therapies), as PDAC is unlikely to be controlled with a monotherapy. Resistance to the various mKRAS-based therapies is another important topic that needs to be addressed. While the exact clinical role of these treatments remains to be determined, we suggest that drugging mKRAS and treating patients with pancreatic cancer more broadly can be approached through an immunological lens.

Outstanding Questions.

  • Given that mKRAS inhibitors increase MHC expression and reduce immunosuppression, does blocking mKRAS pharmacologically lead to the expansion and infiltration of mKRAS-specific T cells? Can these inhibitors synergize with mKRAS vaccines and TCR-based therapies?

  • Can mKRAS-targeted therapies be administered to high risk, but otherwise healthy, individuals to intercept tumorigenesis? Will the efficacy of such therapies and their impact on the immune microenvironment vary in different settings (i.e. PanIN versus invasive carcinoma)?

  • What immunotherapies are best suited to be used in combination with mKRAS-specific treatments in the context of PDAC?

  • What is the best setting for mKRAS vaccines – cancer interception, adjuvant therapy, or advanced disease?

  • How will resistance to anti-mKRAS TCR-based therapies manifest in pancreatic cancer patients?

Highlights.

The KRAS oncogene is mutated in the vast majority of tumors of pancreatic cancer patients and directly promotes tumor initiation and progression as well as an immunosuppressive microenvironment, but until recently mutant KRAS (mKRAS) was considered undruggable.

Novel mKRAS inhibitors have shown therapeutic efficacy in preclinical models and clinical trials for numerous KRAS-mutated cancers. These inhibitors ameliorate the immunosuppression that is characteristic of pancreatic cancer and thus are logical candidates to be tested in combination with immunotherapies.

mKRAS is also immunogenic, and endogenous anti-mKRAS T cells exist. T cells engineered with mKRAS-specific T cell receptors have already shown early clinical promise, and next-generation mKRAS vaccines are being studied.

As this emerging field rapidly develops, we suggest that a significant component to “drugging KRAS” may be immunological.

Acknowledgements

This article was supported by grants from the Parker Institute for Cancer Immunotherapy, Stand Up to Cancer-Lustgarten Foundation, Basser Center for BRCA, and the National Cancer Institute (R01 CA229803) to R.H.V. We also thank members of the R.H.V. laboratory and Drs. Ben Stanger, Samantha Kemp, Adham Bear, Beatriz Carreno, and Gerald Linette (University of Pennsylvania) and Dr. Katelyn Byrne (Oregon Health and Science University) for productive collaborations.

Footnotes

Declaration of interests

R.H.V. has received consulting fees from BMS., is an inventor on patents relating to cancer cellular immunotherapy, cancer vaccines, and KRAS immune epitopes, and receives royalties from Boston Children’s Hospital for a licensed research-only monoclonal antibody. The other author reports no disclosures.

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

References

  • 1.Rahib L, et al. (2014) Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 74, 2913–2921 [DOI] [PubMed] [Google Scholar]
  • 2.Siegel RL, et al. (2023) Cancer statistics, 2023. CA Cancer J Clin 73, 17–48 [DOI] [PubMed] [Google Scholar]
  • 3.Conroy T, et al. (2011) FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364, 1817–1825 [DOI] [PubMed] [Google Scholar]
  • 4.Von Hoff DD, et al. (2013) Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369, 1691–1703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Brahmer JR, et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366, 2455–2465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Royal RE, et al. (2010) Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 33, 828–833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Padron LJ, et al. (2022) Sotigalimab and/or nivolumab with chemotherapy in first-line metastatic pancreatic cancer: clinical and immunologic analyses from the randomized phase 2 PRINCE trial. Nat Med 28, 1167–1177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Moore AR, et al. (2020) RAS-targeted therapies: is the undruggable drugged? Nat Rev Drug Discov 19, 533–552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hosein AN, et al. (2022) Translational advances in pancreatic ductal adenocarcinoma therapy. Nat Cancer 3, 272–286 [DOI] [PubMed] [Google Scholar]
  • 10.Downward J (2003) Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 3, 11–22 [DOI] [PubMed] [Google Scholar]
  • 11.Schreiber RD, et al. (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331, 1565–1570 [DOI] [PubMed] [Google Scholar]
  • 12.Lin JH, et al. (2020) Type 1 conventional dendritic cells are systemically dysregulated early in pancreatic carcinogenesis. J Exp Med 217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Hegde S, et al. (2020) Dendritic Cell Paucity Leads to Dysfunctional Immune Surveillance in Pancreatic Cancer. Cancer Cell 37, 289–307 e289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Clark CE, et al. (2009) Immunosurveillance of pancreatic adenocarcinoma: insights from genetically engineered mouse models of cancer. Cancer Lett 279, 1–7 [DOI] [PubMed] [Google Scholar]
  • 15.Evans RA, et al. (2016) Lack of immunoediting in murine pancreatic cancer reversed with neoantigen. JCI Insight 1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Vonderheide RH (2018) The Immune Revolution: A Case for Priming, Not Checkpoint. Cancer Cell 33, 563–569 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hingorani SR, et al. (2003) Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4, 437–450 [DOI] [PubMed] [Google Scholar]
  • 18.Hingorani SR, et al. (2005) Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 [DOI] [PubMed] [Google Scholar]
  • 19.Clark CE, et al. (2007) Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res 67, 9518–9527 [DOI] [PubMed] [Google Scholar]
  • 20.Pylayeva-Gupta Y, et al. (2012) Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell 21, 836–847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bayne LJ, et al. (2012) Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell 21, 822–835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang Y, et al. (2014) CD4+ T lymphocyte ablation prevents pancreatic carcinogenesis in mice. Cancer Immunol Res 2, 423–435 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Cheng H, et al. (2019) Kras(G12D) mutation contributes to regulatory T cell conversion through activation of the MEK/ERK pathway in pancreatic cancer. Cancer Lett 446, 103–111 [DOI] [PubMed] [Google Scholar]
  • 24.Geng X, et al. (2021) Cancer-Associated Fibroblast (CAF) Heterogeneity and Targeting Therapy of CAFs in Pancreatic Cancer. Front Cell Dev Biol 9, 655152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Velez-Delgado A, et al. (2022) Extrinsic KRAS Signaling Shapes the Pancreatic Microenvironment Through Fibroblast Reprogramming. Cell Mol Gastroenterol Hepatol 13, 1673–1699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Huang H, et al. (2022) Mesothelial cell-derived antigen-presenting cancer-associated fibroblasts induce expansion of regulatory T cells in pancreatic cancer. Cancer Cell 40, 656–673 e657 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Krishnamurty AT, et al. (2022) LRRC15(+) myofibroblasts dictate the stromal setpoint to suppress tumour immunity. Nature 611, 148–154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yan R, et al. (2023) T cell-Mediated Development of Stromal Fibroblasts with an Immune-Enhancing Chemokine Profile. Cancer Immunol Res, OF1–OF11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bailey SR, et al. (2014) Th17 cells in cancer: the ultimate identity crisis. Front Immunol 5, 276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ancrile B, et al. (2007) Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis. Genes Dev 21, 1714–1719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Corcoran RB, et al. (2011) STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Res 71, 5020–5029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.McAllister F, et al. (2014) Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 25, 621–637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dey P, et al. (2020) Oncogenic KRAS-Driven Metabolic Reprogramming in Pancreatic Cancer Cells Utilizes Cytokines from the Tumor Microenvironment. Cancer Discov 10, 608–625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zhang Y, et al. (2017) Epithelial-Myeloid cell crosstalk regulates acinar cell plasticity and pancreatic remodeling in mice. Elife 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ischenko I, et al. (2021) KRAS drives immune evasion in a genetic model of pancreatic cancer. Nat Commun 12, 1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jorgovanovic D, et al. (2020) Roles of IFN-gamma in tumor progression and regression: a review. Biomark Res 8, 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Martinez FO and Gordon S (2014) The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Pan Y, et al. (2020) Tumor-Associated Macrophages in Tumor Immunity. Front Immunol 11, 583084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.McAndrews KM, et al. (2021) Exosome-mediated delivery of CRISPR/Cas9 for targeting of oncogenic Kras(G12D) in pancreatic cancer. Life Sci Alliance 4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Janes MR, et al. (2018) Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 172, 578–589 e517 [DOI] [PubMed] [Google Scholar]
  • 41.Lanman BA, et al. (2020) Discovery of a Covalent Inhibitor of KRAS(G12C) (AMG 510) for the Treatment of Solid Tumors. J Med Chem 63, 52–65 [DOI] [PubMed] [Google Scholar]
  • 42.Hallin J, et al. (2020) The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov 10, 54–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Canon J, et al. (2019) The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 575, 217–223 [DOI] [PubMed] [Google Scholar]
  • 44.Hong DS, et al. (2020) KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med 383, 1207–1217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Skoulidis F, et al. (2021) Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med 384, 2371–2381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ou SI, et al. (2022) First-in-Human Phase I/IB Dose-Finding Study of Adagrasib (MRTX849) in Patients With Advanced KRAS(G12C) Solid Tumors (KRYSTAL-1). J Clin Oncol 40, 2530–2538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Strickler JH, et al. (2023) Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N Engl J Med 388, 33–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Waters AM and Der CJ (2018) KRAS: The Critical Driver and Therapeutic Target for Pancreatic Cancer. Cold Spring Harb Perspect Med 8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kemp SB, et al. (2023) Efficacy of a Small-Molecule Inhibitor of KrasG12D in Immunocompetent Models of Pancreatic Cancer. Cancer Discov 13, 298–311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang X, et al. (2022) Identification of MRTX1133, a Noncovalent, Potent, and Selective KRAS(G12D) Inhibitor. J Med Chem 65, 3123–3133 [DOI] [PubMed] [Google Scholar]
  • 51.Hallin J, et al. (2022) Anti-tumor efficacy of a potent and selective non-covalent KRAS(G12D) inhibitor. Nat Med 28, 2171–2182 [DOI] [PubMed] [Google Scholar]
  • 52.Briere DM, et al. (2021) The KRAS(G12C) Inhibitor MRTX849 Reconditions the Tumor Immune Microenvironment and Sensitizes Tumors to Checkpoint Inhibitor Therapy. Mol Cancer Ther 20, 975–985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mugarza E, et al. (2022) Therapeutic KRAS(G12C) inhibition drives effective interferon-mediated antitumor immunity in immunogenic lung cancers. Sci Adv 8, eabm8780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Brazel D, et al. (2023) CodeBreaK 200: Sotorasib (AMG510) Has Broken the KRAS G12C+ NSCLC Enigma Code. Lung Cancer (Auckl) 14, 31–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Vonderheide RH (2020) CD40 Agonist Antibodies in Cancer Immunotherapy. Annu Rev Med 71, 47–58 [DOI] [PubMed] [Google Scholar]
  • 56.Hattori T, et al. (2023) Creating MHC-Restricted Neoantigens with Covalent Inhibitors That Can Be Targeted by Immune Therapy. Cancer Discov 13, 132–145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhang Z, et al. (2022) A covalent inhibitor of K-Ras(G12C) induces MHC class I presentation of haptenated peptide neoepitopes targetable by immunotherapy. Cancer Cell 40, 1060–1069 e1067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jung S and Schluesener HJ (1991) Human T lymphocytes recognize a peptide of single point-mutated, oncogenic ras proteins. J Exp Med 173, 273–276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fossum B, et al. (1994) p21-ras-peptide-specific T-cell responses in a patient with colorectal cancer. CD4+ and CD8+ T cells recognize a peptide corresponding to a common mutation (13Gly-->Asp). Int J Cancer 56, 40–45 [DOI] [PubMed] [Google Scholar]
  • 60.Kubuschok B, et al. (2006) Naturally occurring T-cell response against mutated p21 ras oncoprotein in pancreatic cancer. Clin Cancer Res 12, 1365–1372 [DOI] [PubMed] [Google Scholar]
  • 61.Gjertsen MK, et al. (1995) Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation. Lancet 346, 1399–1400 [DOI] [PubMed] [Google Scholar]
  • 62.Gjertsen MK, et al. (1997) Cytotoxic CD4+ and CD8+ T lymphocytes, generated by mutant p21-ras (12Val) peptide vaccination of a patient, recognize 12Val-dependent nested epitopes present within the vaccine peptide and kill autologous tumour cells carrying this mutation. Int J Cancer 72, 784–790 [DOI] [PubMed] [Google Scholar]
  • 63.Carbone DP, et al. (2005) Immunization with mutant p53- and K-ras-derived peptides in cancer patients: immune response and clinical outcome. J Clin Oncol 23, 5099–5107 [DOI] [PubMed] [Google Scholar]
  • 64.Gjertsen MK, et al. (2001) Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: Clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer 92, 441–450 [DOI] [PubMed] [Google Scholar]
  • 65.Toubaji A, et al. (2008) Pilot study of mutant ras peptide-based vaccine as an adjuvant treatment in pancreatic and colorectal cancers. Cancer Immunol Immunother 57, 1413–1420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Abou-Alfa GK, et al. (2011) Targeting mutated K-ras in pancreatic adenocarcinoma using an adjuvant vaccine. Am J Clin Oncol 34, 321–325 [DOI] [PubMed] [Google Scholar]
  • 67.Weden S, et al. (2011) Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int J Cancer 128, 1120–1128 [DOI] [PubMed] [Google Scholar]
  • 68.Keenan BP, et al. (2014) A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice. Gastroenterology 146, 1784–1794 e1786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rojas LA, et al. (2023) Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature 618, 144–150 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang QJ, et al. (2016) Identification of T-cell Receptors Targeting KRAS-Mutated Human Tumors. Cancer Immunol Res 4, 204–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bear AS, et al. (2021) Biochemical and functional characterization of mutant KRAS epitopes validates this oncoprotein for immunological targeting. Nat Commun 12, 4365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Choi J, et al. (2021) Systematic discovery and validation of T cell targets directed against oncogenic KRAS mutations. Cell Rep Methods 1, 100084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Levin N, et al. (2021) Identification and Validation of T-cell Receptors Targeting RAS Hotspot Mutations in Human Cancers for Use in Cell-based Immunotherapy. Clin Cancer Res 27, 5084–5095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Poole A, et al. (2022) Therapeutic high affinity T cell receptor targeting a KRAS(G12D) cancer neoantigen. Nat Commun 13, 5333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Douglass J, et al. (2021) Bispecific antibodies targeting mutant RAS neoantigens. Sci Immunol 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tran E, et al. (2016) T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer. N Engl J Med 375, 2255–2262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Leidner R, et al. (2022) Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. N Engl J Med 386, 2112–2119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Schultze JL and Vonderheide RH (2001) From cancer genomics to cancer immunotherapy: toward second-generation tumor antigens. Trends Immunol 22, 516–523 [DOI] [PubMed] [Google Scholar]
  • 79.Ryschich E, et al. (2005) Control of T-cell-mediated immune response by HLA class I in human pancreatic carcinoma. Clin Cancer Res 11, 498–504 [PubMed] [Google Scholar]
  • 80.Pandha H, et al. (2007) Loss of expression of antigen-presenting molecules in human pancreatic cancer and pancreatic cancer cell lines. Clin Exp Immunol 148, 127–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Yamamoto K, et al. (2020) Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 581, 100–105 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES