How do different drug classes work in treating Stomach Cancer?

17 March 2025

Overview of Stomach Cancer

Stomach cancer, most commonly in the form of gastric adenocarcinoma, is a major global health problem that affects millions of people worldwide. Gastric cancer comprises several histological subtypes that reflect differing degrees of differentiation and biological behavior. The disease is recognized not only for its heterogeneous nature but also for its highly variable clinical presentation. The pathological types include differentiated-type adenocarcinoma, poorly differentiated adenocarcinoma, mucinous adenocarcinoma, and several other rare forms that may also be seen depending on the tumor’s underlying molecular and genetic profile.

Definition and Types

Gastric cancer is defined as a malignant neoplasm arising from the epithelial lining of the stomach. It often presents as an adenocarcinoma, which may then be further subdivided based on histomorphology into categories such as intestinal and diffuse types according to Lauren’s classification. In addition to these broad classifications, several studies have shown that additional subtypes exist—for example, the refined categorization into differentiated versus poorly differentiated types that correlates with progression from precancerous conditions like intestinal metaplasia toward a more aggressive phenotype. In particular, the presence of intestinal epithelial metaplasia is seen as a precursor lesion in the differentiated type and suggests a stepwise progression in tumorigenesis. This heterogeneity is critical for treatment strategies since the molecular drivers and prognosis vary significantly among types.

Epidemiology and Risk Factors

Gastric cancer is the fifth most common cancer and the fourth leading cause of cancer-related death on a global scale. Although incidence rates have declined in some parts of the world, they remain high in regions such as East Asia due to dietary factors, Helicobacter pylori infection, high salt intake, tobacco use, and genetic predispositions. Epidemiological studies confirm that environmental exposures in combination with genetic susceptibility play a critical role. In addition, factors such as obesity, pernicious anemia, and a history of partial gastrectomy are also linked with higher risk. The variability in risk factors among populations underscores why clinical outcomes and response to various therapeutic regimens may differ across ethnic groups and geographic regions.

Drug Classes Used in Stomach Cancer Treatment

Successful treatment of gastric cancer generally requires a multimodal approach. Over the recent decades, three major drug classes have emerged to combat the disease: chemotherapy agents, targeted therapy drugs, and immunotherapy options. Each class employs a distinct mechanism of action and is chosen based on factors such as tumor stage, molecular profile, patient performance status, and anticipated adverse events.

Chemotherapy Agents

Chemotherapy represents the long‐established foundation for systemic treatment of advanced stomach cancer. Classic chemotherapeutic regimens include cytotoxic agents such as fluoropyrimidines (5-fluorouracil, capecitabine, and S-1), platinum-based compounds (cisplatin and oxaliplatin), taxanes (docetaxel and paclitaxel), and topoisomerase inhibitors (irinotecan). These drugs have been combined as doublet or triplet regimens to maximize the impact on tumor growth while attempting to provide a balance between efficacy and tolerability. The benefits of these agents have been affirmed in multiple clinical trials that compared combination regimens versus monotherapy, demonstrating significant improvement in response rate, progression-free survival (PFS), and overall survival (OS). Despite their success, the modest overall improvements underscore the need for precision and personalization in chemotherapy treatments due to the heterogeneity of both tumor biology and patient health status.

Targeted Therapy Drugs

Over recent years, targeted therapies have emerged as a powerful paradigm shift in the treatment of gastric cancer. These drugs specifically interact with molecular abnormalities critical for tumor survival and proliferation. For example, trastuzumab, a monoclonal antibody targeting the HER2 receptor, is used in HER2-positive gastric tumors, thereby blocking downstream proliferative signals. Other targeted agents include ramucirumab, an anti–vascular endothelial growth factor receptor-2 (VEGFR2) antibody, which impedes angiogenesis—a vital process for tumor blood supply. In addition, small molecule inhibitors acting on receptor tyrosine kinases (RTKs) and the intracellular signaling pathways are increasingly being integrated into treatment strategies. These therapies are typically administered either as single agents or, more commonly, in conjunction with conventional chemotherapy to harness their synergistic effects.

Immunotherapy Options

Immunotherapy represents one of the most exciting and rapidly evolving areas in cancer treatment. In the context of stomach cancer, immune checkpoint inhibitors (ICIs) such as nivolumab, pembrolizumab, and others have been investigated extensively. These agents work by reactivating the body’s endogenous immune responses against tumor cells, taking advantage of the concept of tumor “immune escape.” Other immunotherapeutic approaches include therapeutic cancer vaccines, adoptive cell transfer (such as chimeric antigen receptor T cell therapy, CAR-T), and strategies aimed at targeting the immunosuppressive tumor microenvironment. Although immunotherapy alone has produced durable responses in a subset of patients—especially those with high PD-L1 expression or microsatellite instability—it is often used in combination with chemotherapy or targeted therapy to further augment clinical benefits.

Mechanisms of Action

A detailed understanding of how each drug class exerts its effects on tumor cells is fundamental for rational therapy selection. The distinct mechanisms of action provide the foundation for their clinical application and help explain the observed efficacy and patterns of resistance that frequently challenge clinical outcomes.

How Chemotherapy Works

Chemotherapy agents generally act by damaging or interfering with cellular processes that are essential for tumor cell proliferation. For example, 5-FU and its orally available analogs capecitabine and S-1 are antimetabolites that interrupt DNA synthesis by inhibiting thymidine synthase. This causes disruptions during the DNA replication phase, leading to apoptosis in rapidly dividing tumor cells. Platinum-based compounds such as cisplatin and oxaliplatin form covalent bonds with DNA to create intra- and inter-strand crosslinks, thereby stalling the DNA replication machinery and ultimately triggering cell death. Additionally, taxanes such as docetaxel and paclitaxel target microtubules—one of the essential structures required for cell division. They stabilize the microtubule polymer and prevent its depolymerization, which leads to mitotic arrest and subsequent apoptosis. Irinotecan, an inhibitor of topoisomerase I, prevents the resealing of single-strand breaks in DNA that occur during replication, resulting in lethal double-strand DNA damage. Although these cytotoxic agents are nonselective and affect normal proliferating cells as well, their rapid effect on tumor growth helps reduce tumor burden and provides palliation.

Mechanism of Targeted Therapy

Targeted therapy drugs are designed to interfere with specific molecules or signaling pathways that are aberrantly activated in gastric cancer cells. For instance, trastuzumab binds to the extracellular domain of the HER2 protein, which is overexpressed in a subset of gastric cancers. This binding blocks HER2-mediated signaling, including the PI3K/AKT and MAPK pathways, that would otherwise promote tumor cell growth and survival. Ramucirumab, on the other hand, binds to VEGFR2, thereby inhibiting the vascular endothelial growth factor (VEGF) driven angiogenesis that is critical for tumor growth and metastasis. In addition to these monoclonal antibodies, small molecule inhibitors target intracellular tyrosine kinase domains, preventing signal transduction from receptors even when ligand binding occurs. The highly selective nature of these agents allows for relatively lower off-target toxicity, although their effectiveness is largely dependent on the molecular characteristics of the tumor. Resistance mechanisms, such as mutations in the binding domain, reactivation of bypass signaling pathways, and tumor heterogeneity, still pose major challenges; however, the identification of predictive biomarkers such as HER2 amplification or MET gene amplification has improved patient selection.

Action of Immunotherapy

Immunotherapy harnesses the power of the patient’s own immune system to combat tumor cells. The cornerstone of many immunotherapeutic strategies in gastric cancer is immune checkpoint blockade. In a normal physiological state, checkpoint molecules such as PD-1 on T cells and PD-L1 on tumor cells function to maintain self-tolerance and prevent autoimmunity. However, tumor cells frequently exploit these pathways by upregulating PD-L1 to suppress immune attack. Immune checkpoint inhibitors, such as pembrolizumab and nivolumab, block these interactions, thereby reactivating T cell function and enabling an immune-mediated tumor cell kill. In addition, other forms of immunotherapy, including cancer vaccines and adoptive cell transfer, aim to prime or expand populations of tumor-specific T cells. Cancer vaccines introduce tumor-associated antigens to the immune system to promote an adaptive immune response, while adoptive cell therapies involve the ex vivo expansion and reinfusion of autologous T lymphocytes that are engineered to recognize specific tumor antigens. A key advantage in these approaches is the establishment of immune memory, which can result in long-term protection and durable clinical responses even after cessation of treatment.

Clinical Outcomes and Effectiveness

The clinical efficacy of the different drug classes used in stomach cancer treatment is influenced by the distinct mechanisms of action and the biological heterogeneity of gastric cancer. Comparative studies and clinical trials have provided insights into the outcomes associated with each modality, highlighting both successes and areas in need of further improvement.

Efficacy of Different Drug Classes

Chemotherapy remains a mainstay treatment, particularly for advanced gastric cancer, and has been shown to improve overall survival and disease control compared to best supportive care alone. Combination chemotherapy regimens, such as those incorporating fluoropyrimidines with a platinum compound and taxanes, have yielded higher response rates and prolonged progression-free survival. However, the improvements, while statistically significant, are sometimes modest, and drug resistance remains a frequent challenge.

Targeted therapies have brought a new era of precision medicine to gastric cancer. For instance, trastuzumab in combination with chemotherapy has led to improved survival in patients with HER2-positive tumors. Similarly, ramucirumab has demonstrated benefit when used either as monotherapy or in combination with paclitaxel in patients who have progressed on first-line therapy. The efficacy of small molecule inhibitors is more variable and often depends on the specific driver mutations present in the tumor. Despite these advances, only a subset of patients exhibit significant responses to targeted therapies, underscoring the need for robust biomarker-based patient selection.

Immunotherapy, particularly checkpoint inhibitors, has shown promising durable responses in a fraction of gastric cancer patients. For example, clinical trials such as CheckMate 649 have demonstrated overall survival benefits for patients treated with nivolumab combined with chemotherapy, especially in populations with high PD-L1 expression and microsatellite instability. However, the response rate to immunotherapy tends to be lower when applied as a single agent, and many patients may not achieve meaningful clinical benefit unless immunotherapy is combined with chemotherapy or other agents.

Case Studies and Clinical Trials

Multiple clinical trials provide concrete evidence for the efficacy of these therapeutic modalities. The FLOT regimen (a taxane-based triplet involving fluorouracil, leucovorin, oxaliplatin, and docetaxel) has been evaluated against other regimens such as the SOX regimen (S-1 plus oxaliplatin) in randomized clinical trials. These studies indicate that taxane-containing triplet regimens may result in improved pathological response and longer progression-free intervals, although toxicity profiles vary between regimens. Other pivotal trials such as the ToGA study have firmly established the role of HER2-targeted therapy with trastuzumab for HER2-positive gastric cancer, leading to significant improvements in survival when combined with chemotherapy. Moreover, the CheckMate 649 and ATTRACTION-4 studies have elucidated the potential benefit of immune checkpoint inhibitors, either as monotherapy or in combination with chemotherapy, with results indicating subgroup benefits related to PD-L1 expression levels. Recent network meta-analyses comparing various chemotherapy regimens have further highlighted the relative benefits and adverse event profiles across different drug combinations, emphasizing that regimens incorporating capecitabine or S-1 along with platinum compounds may yield better disease control rates.

Challenges and Future Directions

Although considerable progress has been made in the pharmacologic treatment of gastric cancer, significant challenges remain. The intrinsic heterogeneity of the tumor, therapy-induced resistance, and treatment-related toxicity have presented ongoing hurdles in achieving long-term disease control. Researchers continue to explore and develop new strategies to overcome these limitations, with the hope of further improving survival outcomes and patient quality of life.

Current Limitations

One of the foremost challenges in the treatment of stomach cancer is the development of drug resistance. Even when initial responses are encouraging, tumors frequently acquire resistance through various mechanisms, such as enhanced drug efflux, alterations in drug target modifications, activation of compensatory signaling pathways, and changes in the tumor microenvironment. For instance, with conventional chemotherapy, multidrug resistance often limits the duration of response, necessitating the development of combination strategies and rotational regimens. Similarly, targeted therapies may be rendered ineffective if mutations develop in the target protein or if alternative growth pathways compensate for the blocked signaling.

Furthermore, immunotherapies, despite showing durable responses in some patients, face the challenge of a “cold” tumor microenvironment in many gastric cancers, where there is minimal T cell infiltration and hence a suboptimal immune response. Many clinical trials have reported limited benefit in unselected patient populations, underscoring the need for predictive biomarkers to identify patients who will most likely respond. Safety is another significant concern across all drug classes. Chemotherapy’s non-specific toxicity can result in severe adverse effects, and although targeted therapies tend to have a more favorable side effect profile, they are not devoid of adverse events that may lead to treatment discontinuation. Additionally, immune-related adverse events from checkpoint inhibitors—though generally less cytotoxic—can occasionally be severe and unpredictable.

Emerging Therapies and Research

In response to these challenges, extensive research is focused on emerging therapies and novel management strategies for gastric cancer. One promising area is the integration of multiple therapeutic modalities to maximize antitumor efficacy while minimizing toxicity. For example, combining immunotherapy with chemotherapy or targeted therapies may help convert a “cold” tumor microenvironment into a “hot” one, thereby augmenting the immune response. Concurrently, the exploration of newer biomarkers, including microRNAs, circulating tumor DNA, and next-generation sequencing approaches, is enabling a more precision-based treatment selection that tailors therapy to the individual tumor’s genetic profile.

Recent advancements include approaches for rapidly assessing drug sensitivity using nucleic acid markers, which might eventually allow clinicians to predict therapeutic response early in the treatment course and adjust therapies in real time. In addition, patents have been filed for innovative methods that combine targeted antibodies with conventional chemotherapies in novel dosing and administration schedules, aiming to potentiate efficacy and overcome drug resistance. Researchers are also evaluating the utility of proteomics in identifying drug resistance mechanisms, which could lead to the development of combination therapies that target multiple resistance pathways simultaneously.

Furthermore, clinical trial designs are evolving to better capture the benefits of these emerging therapies. Adaptive trial designs and umbrella trials, where multiple therapeutic agents are tested within a single framework, are beginning to address the inherent heterogeneity seen in gastric cancer populations while expediting the evaluation of novel combinations. These innovative designs allow for dynamic assessment and modification of treatment regimens based on interim results, thereby shortening the time needed to identify effective therapies and improving overall trial outcomes.

The future of stomach cancer treatment will likely depend on integrated strategies that encompass chemotherapy, targeted therapy, and immunotherapy—each playing a complementary role. There is an ongoing focus on developing highly selective targeted agents that have minimal off-target toxicity while preserving robust antitumor activity, as well as on immunomodulatory agents that can overcome the immunosuppressive tumor microenvironment. The integration of advanced diagnostic tools to monitor molecular changes in real time holds promise for truly personalized treatment regimens. Such approaches are expected to not only extend overall survival but also maintain quality of life by minimizing adverse events and facilitating long-term disease control.

In summary, clinicians are increasingly adopting a multimodal strategy wherein traditional cytotoxic agents are combined with biologically targeted and immunomodulatory approaches. This approach is supported by emerging data from randomized clinical trials and meta-analyses showing improved pathological response rates and prolongation of survival in selected patient populations. The ability to dynamically modify therapy based on early biomarkers and genetic profiling may eventually lead to therapies that are both more effective and less toxic than current standards.

Conclusion

In conclusion, the treatment of stomach cancer involves a multifaceted approach that utilizes distinct drug classes—chemotherapy, targeted therapy, and immunotherapy—each working through different mechanisms of action. Chemotherapy exploits the rapid cell division of tumor cells by damaging DNA synthesis, interfering with microtubule dynamics, and inhibiting essential enzymes required for cell replication. Targeted therapies focus on inhibiting specific signaling pathways, such as HER2 and VEGFR2 pathways, that are crucial for tumor growth and angiogenesis. Immunotherapy, on the other hand, works by reactivating the body’s immune system, primarily by blocking inhibitory checkpoint molecules like PD-1 and PD-L1, thereby unleashing T-cell–mediated tumor destruction.

These drug classes have demonstrated varying degrees of efficacy. Chemotherapy, though effective in reducing tumor bulk and extending survival compared with supportive care, is often limited by cytotoxicity and resistance mechanisms. Targeted therapies provide a more personalized approach; their effectiveness is largely determined by the molecular characteristics of the tumor, such as HER2 amplification, and have shown significant survival benefits when combined with chemotherapy. Immunotherapy has heralded a new era in cancer treatment with its potential for durable responses and immune memory, though its success is contingent on overcoming the suppressive tumor microenvironment and requires careful patient selection based on biomarkers like PD-L1 expression.

Clinical trial data from studies such as the FLOT versus SOX trials, the ToGA study, CheckMate 649, and others underscore the potential and challenges of these strategies. While combination regimens have often improved outcomes, drug resistance and adverse effects remain significant hurdles. Current limitations include the development of primary and acquired drug resistance, toxicity profiles, and the challenge of patient selection due to tumor heterogeneity. In response, emerging therapies and research are focused on integrating multiple modalities, employing adaptive clinical trial designs, and using advanced molecular diagnostics to individualize treatment plans. Innovative approaches, such as rapid drug sensitivity assessments, proteomics for resistance marker detection, and the development of novel combination formulations, offer promise for further advancements in the field.

The future of gastric cancer treatment lies in a comprehensive, multimodal approach that aligns with the principles of precision medicine. By combining cytotoxic, targeted, and immune-based therapies—and fine-tuning these interventions based on detailed molecular profiling—it may be possible to overcome current therapeutic limitations and improve both survival rates and quality of life for patients with stomach cancer. This integrative strategy is supported by evolving clinical trial evidence and has the potential to fundamentally change the prognosis of a historically challenging disease. In essence, the diverse mechanisms of action across drug classes not only provide multiple angles to attack tumor biology but also create opportunities for synergistic effects that may ultimately lead to more effective and durable treatment outcomes.

Overall, while each drug class works through distinct biological mechanisms—from direct cytotoxic effects to blockade of specific signaling pathways and restoration of antitumor immunity—the ultimate goal remains the same: to halt tumor progression, overcome resistance, and improve patient survival. The journey from conventional chemotherapy to sophisticated targeted and immunotherapies represents not only a paradigm shift in the treatment of gastric cancer but also a promising roadmap toward personalized medicine. Future research, adaptive clinical trial designs, and integrative diagnostic tools will be crucial in transforming these emerging strategies into standard clinical practice, ultimately ensuring that gastric cancer treatment continues to evolve and deliver better outcomes for patients worldwide.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成