Recent discoveries of new therapeutic targets within recent research are driving the development of innovative combinatorial therapies, while concurrently deepening our understanding of several distinct cell death pathways. NB 598 nmr These methods, although successful in reducing the therapeutic threshold, still present a significant concern regarding the potential for subsequent resistance to emerge. The basis for future PDAC treatments, free from excessive health risks, may be found in the discovery of resistance-targeting approaches, used alone or together. We investigate the factors contributing to PDAC chemoresistance in this chapter, and explore countermeasures targeting various pathways and cellular functions involved in the development and sustenance of chemoresistance.
Pancreatic ductal adenocarcinoma (PDAC) is the most common form of pancreatic neoplasm, comprising 90% of cases, and remains one of the most lethal cancers among all malignancies. Aberrant oncogenic signaling, harbored by PDAC, potentially originates from diverse genetic and epigenetic modifications, including driver gene mutations (KRAS, CDKN2A, p53), regulatory gene amplifications (MYC, IGF2BP2, ROIK3), and disruptions in chromatin-modifying proteins (HDAC, WDR5), among other factors. The occurrence of Pancreatic Intraepithelial Neoplasia (PanIN), a significant event, is frequently attributed to activating mutations within the KRAS gene. KRAS mutations can orchestrate a range of signaling pathways, influencing subsequent targets like MYC, significantly contributing to the advancement of cancer. Major oncogenic signaling pathways are explored in this review, drawing on recent research to understand the genesis of PDAC. Epigenetic reprogramming and metastasis are shown to be significantly affected by MYC, both directly and indirectly through its interaction with KRAS. Moreover, a summary of recent single-cell genomic research findings is presented, emphasizing the variability observed in pancreatic ductal adenocarcinoma (PDAC) and its tumor microenvironment, thereby suggesting molecular targets for future PDAC therapies.
A diagnosis of pancreatic ductal adenocarcinoma (PDAC) is frequently delayed due to the disease's typically advanced or metastatic presentation. The United States projects a rise of 62,210 new cases and 49,830 deaths by the conclusion of this year, with an overwhelming 90% being linked to the PDAC subtype. Despite improvements in cancer treatment, the diverse nature of pancreatic ductal adenocarcinoma (PDAC) tumors, both between patients and within the same patient's primary and metastatic lesions, continues to pose a substantial obstacle to its successful eradication. extra-intestinal microbiome The review examines PDAC subtypes, drawing upon genomic, transcriptional, epigenetic, and metabolic markers found in patient samples and individual tumor specimens. Studies in PDAC biology, conducted recently, suggest that PDAC heterogeneity, operating under stress conditions such as hypoxia and nutrient deprivation, significantly impacts disease progression and results in metabolic reprogramming. Hence, we broaden our insight into the root causes that impede the interaction between extracellular matrix components and tumor cells, ultimately shaping the mechanics of tumor growth and metastasis. The bilateral relationship between pancreatic ductal adenocarcinoma (PDAC) cells and the heterogeneous tumor microenvironment's components plays a crucial role in determining the tumor's growth potential and response to therapy, thus providing an avenue for successful therapeutic approaches. Furthermore, the dynamic exchange between stromal and immune cells significantly affects the immune response, including surveillance or evasion, and thereby influences the intricate process of tumor formation. In a nutshell, the review consolidates current information about PDAC treatments, focusing on the multifaceted nature of tumor heterogeneity, which affects disease progression and treatment response in the face of stress.
Minority patients with pancreatic cancer, often underrepresented, experience varied access to cancer treatments, including clinical trials. In order to enhance outcomes for individuals with pancreatic cancer, the completion and successful execution of clinical trials is of utmost importance. Consequently, careful consideration must be given to the optimal utilization of strategies to maximize patient eligibility for both therapeutic and non-therapeutic clinical trials. Recognition of the obstacles, individual, clinician, and system-level, that hinder clinical trial recruitment, enrollment, and completion, is paramount for clinicians and the health system to combat bias. For cancer clinical trials to yield generalizable results and advance health equity, strategies focused on increasing enrollment among underrepresented minorities, socioeconomically disadvantaged individuals, and underserved communities are essential.
In human pancreatic cancer, KRAS, a key player in the RAS family of genes, is the most frequently mutated oncogene, appearing in ninety-five percent of cases. KRAS mutations cause a continuous state of activation, thereby activating downstream signaling pathways, such as RAF/MEK/ERK and PI3K/AKT/mTOR, which results in the proliferation of cells and the prevention of programmed cell death in cancer cells. The development of the first covalent inhibitor, focused on the G12C mutation in KRAS, demonstrated that what was once considered 'undruggable' was indeed treatable. G12C mutations, prevalent in non-small cell lung cancer, appear far less common in pancreatic cancer. Conversely, pancreatic cancer often presents with alternative KRAS mutations, including G12D and G12V. In contrast to the existing inhibitors for other mutations, recent developments include inhibitors targeting the G12D mutation, including MRTX1133. Cultural medicine Unfortunately, KRAS inhibitor monotherapy's therapeutic impact is thwarted by the development of resistance. Accordingly, multiple methods of combining therapies were tested, and some generated encouraging outcomes, including combinations that used receptor tyrosine kinase, SHP2, or SOS1 inhibitors. Our investigation recently highlighted a synergistic effect of sotorasib combined with DT2216, a BCL-XL-selective degrader, on the inhibition of G12C-mutated pancreatic cancer cell proliferation, evident in both laboratory and animal testing. Resistance to KRAS-targeted therapies is partly due to their effect on inducing cell cycle arrest and cellular senescence. The combination of these therapies with DT2216, however, results in a more effective induction of apoptosis. Combinatorial approaches, structurally similar to those used elsewhere, could have positive effects on G12D inhibitors in pancreatic cancer. This chapter will investigate KRAS biochemistry, its signaling pathways, the various forms of KRAS mutations, innovative KRAS-targeted therapeutic approaches, and strategies for combining these approaches. We conclude by examining the difficulties of KRAS inhibition, specifically in pancreatic cancer, and outline emerging future directions.
Pancreatic Ductal Adenocarcinoma, or PDAC, a frequently aggressive form of pancreatic cancer, is typically diagnosed at a late stage, often hindering treatment options and leading to limited clinical responses. According to projected figures for 2030, pancreatic ductal adenocarcinoma is anticipated to be the second leading cause of cancer-related deaths in the United States. Drug resistance poses a significant obstacle to the overall survival of patients with pancreatic ductal adenocarcinoma (PDAC). In pancreatic ductal adenocarcinoma (PDAC), virtually all cases (over 90%) exhibit a consistent pattern of oncogenic KRAS mutations. Unfortunately, the clinical application of drugs specifically designed to address frequent KRAS mutations in pancreatic cancer remains unavailable. For this reason, the research into alternative druggable targets or treatment strategies to improve patient care persists in the context of pancreatic ductal adenocarcinoma. In the majority of pancreatic ductal adenocarcinoma (PDAC) instances, KRAS mutations activate the RAF-MEK-MAPK pathways, thereby initiating pancreatic tumor development. The pancreatic cancer tumor microenvironment (TME) and chemotherapy resistance are profoundly influenced by the MAPK signaling cascade (MAP4KMAP3KMAP2KMAPK). In pancreatic cancer, the immunosuppressive tumor microenvironment (TME) presents a further barrier to the successful therapy using chemotherapy and immunotherapy. The immune checkpoint proteins CTLA-4, PD-1, PD-L1, and PD-L2 are key contributors to the interplay between T cell dysfunction and pancreatic tumor cell growth. Here, we comprehensively review the activation of MAPKs, a molecular characteristic of KRAS mutations, and its influence on the pancreatic cancer tumor microenvironment, chemoresistance to treatment, and the expression of immune checkpoint proteins, factors which might impact clinical outcomes in pancreatic ductal adenocarcinoma. Thus, elucidating the dynamic interaction between MAPK signaling cascades and the tumor microenvironment (TME) holds the key to designing more effective combined therapies employing both immunotherapy and MAPK inhibitors for pancreatic cancer.
The Notch signaling pathway, a conserved signaling cascade crucial for both embryonic and postnatal development, is, however, also linked to tumorigenesis in numerous organs, including the pancreas, when aberrant. Unfortunately, pancreatic ductal adenocarcinoma (PDAC), the most frequent malignancy of the pancreas, displays unacceptably low survival rates stemming from late diagnoses and its specific resistance to therapies. Preneoplastic lesions and PDACs, in genetically engineered mouse models and human patients, exhibit upregulation of the Notch signaling pathway. Conversely, Notch signaling inhibition effectively suppresses tumor development and progression in mice and patient-derived xenograft tumor growth, emphasizing Notch's critical role in PDAC. However, the significance of the Notch signaling pathway in pancreatic ductal adenocarcinoma is still unclear, exemplified by the diverse functions of Notch receptors and the contrasting consequences of inhibiting Notch signaling in murine models of PDAC that stem from different cellular origins or are examined at disparate stages.