How do different drug classes work in treating Melanoma?

17 March 2025
Overview of Melanoma
Definition and Epidemiology
Melanoma is a type of skin cancer that arises from melanocytes—the pigment‐producing cells found primarily in the basal layer of the epidermis. It is widely regarded as the most aggressive and deadly form of skin malignancy due to its inherent ability to metastasize early and its resistance to many conventional therapies. Epidemiological studies consistently show that melanoma incidence is rising across the globe with particularly high rates in fair‐skinned populations, where ultraviolet (UV) radiation exposure plays a critical role in disease pathogenesis. Recent global data indicate that although melanoma accounts for only a small percentage of all skin cancers, it is responsible for the vast majority of skin cancer–related deaths. For instance, nearly 100,000 new cases were predicted in the U.S. in 2023 with significant mortality numbers, especially for advanced or metastatic disease stages. In addition, melanoma is associated with genetic predisposition, and its incidence is influenced by multiple factors ranging from UV exposure, incidence of atypical nevi, personal and family history to lifestyle factors.

Current Treatment Landscape
Historically, melanoma was managed with a combination of surgical excision for early‐stage disease and conventional chemotherapeutic regimens for advanced cases. However, the introduction of newer agents in the past two decades has revolutionized the treatment paradigm. Today, along with surgical management and radiotherapy, systemic treatments predominantly include immunotherapy and targeted therapy. Immunotherapy—most notably immune checkpoint inhibitors—has led to significant improvements in long‐term survival for advanced melanoma patients, while targeted agents such as BRAF and MEK inhibitors are used in patients with specific gene mutations that drive tumor growth. Chemotherapy still plays a role, especially in settings where other modalities are inaccessible or ineffective, though its impact on long‐term outcomes is generally modest. This evolving landscape is now characterized by rational combinations of therapies to overcome treatment resistance, minimize adverse effects, and ultimately improve overall prognosis.

Drug Classes Used in Melanoma Treatment
Immunotherapy
Immunotherapy represents a rapid shift from non-specific immune stimulation to precise modulation of the immune system. The most established immunotherapeutic strategies, particularly for melanoma, include immune checkpoint inhibitors such as anti–CTLA-4 antibodies (e.g., ipilimumab) and anti–PD-1 agents (e.g., nivolumab and pembrolizumab). These drugs work by blocking inhibitory signals that normally downregulate T-cell activity, thereby reactivating the immune system against tumor cells. Beyond these, approaches like interleukin-2 (IL-2) therapy and adoptive T-cell transfer—where tumor-infiltrating lymphocytes are expanded ex vivo and reinfused—have also historically contributed to melanoma treatment, albeit with more limited utility due to toxicity and variable efficacy. More novel strategies such as anti-LAG-3 antibodies in combination with PD-1 blockade further expand the immunotherapeutic armamentarium, with promising early clinical data that suggest improved outcomes, although these are still under investigation.

Targeted Therapy
Targeted therapies for melanoma focus on inhibiting pivotal molecular pathways that drive tumor cell proliferation and survival. The discovery that approximately 40–50% of cutaneous melanomas harbored a BRAF mutation—most commonly the V600E variant—sparked the development of BRAF inhibitors such as vemurafenib and dabrafenib. These agents act by selectively inhibiting the BRAF kinase activity, thereby disrupting the downstream MAPK signaling cascade that is critical for melanoma cell growth and survival. Combined targeted strategies, wherein a BRAF inhibitor is paired with a MEK inhibitor (e.g., trametinib or cobimetinib), have become the standard of care for patients with BRAF-mutated melanoma. This combination not only enhances efficacy by further inhibiting the MAPK pathway but also delays the onset of resistance by mitigating paradoxical activation of the pathway that is sometimes seen with BRAF inhibitors alone. Other molecular targets include components of the PI3K/AKT pathway, and research into novel compounds that can modulate these pathways continues to evolve, aiming to overcome resistance mechanisms and enhance treatment durability.

Chemotherapy
Chemotherapy comprises the more traditional cytotoxic agents that have been used for decades in patients with advanced melanoma, especially those with refractory disease or when other options are not feasible. Agents such as dacarbazine (DTIC) and temozolomide work by damaging the DNA of rapidly dividing cells through various mechanisms, including alkylation and inhibition of DNA replication. Despite being the first systemic treatment modalities approved for melanoma, chemotherapy generally results in low objective response rates and limited overall survival benefits—often less than 12% response rates—with many patients developing intrinsic or acquired chemoresistance. This reduced effectiveness is partly due to the high degree of genetic heterogeneity and robust anti-apoptotic mechanisms that are characteristic of melanoma cells. Additionally, chemotherapy is associated with a broad toxicity profile that affects both tumor cells and normal proliferating cells, leading to significant adverse events such as myelosuppression, nausea, vomiting, and organ toxicities.

Mechanisms of Action
How Immunotherapy Works
Immunotherapy harnesses the body's natural defense mechanisms by modulating the immune system to recognize and attack melanoma cells. The fundamental mechanism involves blocking immune checkpoint proteins—such as CTLA-4 and PD-1—that normally act as brakes to temper immune responses. Under healthy conditions, these checkpoints prevent autoimmunity by limiting overactivation of T cells; however, melanoma cells often exploit this system by inducing an immunosuppressive microenvironment that hinders effective T-cell–mediated tumor elimination.
When anti–CTLA-4 antibodies like ipilimumab are administered, they block the CTLA-4 receptor on T cells and prevent it from binding to its ligands (CD80/CD86) on antigen-presenting cells. This blockade results in enhanced T-cell activation and proliferation and allows the immune system to mount a stronger cytotoxic attack on tumor cells. Similarly, PD-1 inhibitors—nivolumab and pembrolizumab—work by preventing PD-1 on T cells from interacting with PD-L1 expressed on melanoma cells or tumor-associated immune cells. This disruption lifts the inhibition on T cells within the tumor microenvironment, thereby enabling them to vigorously attack the malignant cells.
Other immunotherapeutic approaches include adoptive cell therapy, where T cells extracted from tumor samples (tumor-infiltrating lymphocytes) are expanded outside the body and reinfused into the patient. Such personalized immunotherapy is designed to boost the number of potent anti–tumor T cells and has shown promising clinical responses in selected patient populations, though it requires complex processing and monitoring. Overall, by reversing immune tolerance and enabling a robust immune response, immunotherapy has provided durable responses and improved survival outcomes for many patients with melanoma, albeit sometimes at the price of immune-mediated adverse events affecting multiple organ systems.

How Targeted Therapy Works
Targeted therapy in melanoma is designed to interfere with specific molecular alterations that drive melanoma carcinogenesis. A prime example is the inhibition of the MAPK pathway, which is aberrantly activated in a substantial proportion of melanomas due to mutations in the BRAF gene. In patients harboring these mutations, drugs like vemurafenib and dabrafenib bind to the mutant BRAF kinase and inhibit its activity, leading to decreased phosphorylation of downstream MEK and ERK proteins that are essential for cell proliferation and survival.
However, monotherapy with BRAF inhibitors is often limited by the development of resistance as tumor cells adapt through reactivation of the MAPK pathway or through parallel survival pathways. To combat this, clinicians now frequently use combination therapy: for example, pairing a BRAF inhibitor with a MEK inhibitor (trametinib or cobimetinib) results in a more complete blockade of the MAPK cascade, leads to deeper tumor responses, and delays the emergence of resistance mechanisms.
Furthermore, targeted therapy can extend its reach to other oncogenic pathways. The PI3K/AKT/mTOR pathway, which also plays a pivotal role in cell growth, survival, and metabolism, is another target under investigation. Interrupting these signaling cascades disrupts cellular homeostasis, ultimately inducing apoptosis of melanoma cells. The specificity of targeted therapies allows for rapid tumor shrinkage and measurable improvements in progression-free survival, although acquired resistance remains a clinical challenge that often necessitates additional or sequential therapies.

How Chemotherapy Works
Chemotherapy employs cytotoxic drugs that disrupt cellular processes required for cell division. In melanoma, agents such as dacarbazine (DTIC) and temozolomide work chiefly by alkylating DNA bases, which leads to the formation of DNA adducts and cross-links and ultimately results in the interruption of DNA replication and transcription. This DNA damage can trigger cell cycle arrest and apoptosis particularly in rapidly dividing cells.
Despite these mechanisms, melanoma has proven to be particularly resistant to chemotherapy. Several factors contribute to this resistance: the efficient DNA repair systems within melanoma cells, the overexpression of anti-apoptotic proteins that prevent programmed cell death, and the presence of multidrug resistance (MDR) pumps that actively efflux chemotherapeutic agents from the tumor cells. The therapeutic index of chemotherapy is further hampered by the fact that these drugs do not differentiate well between malignant and healthy cells. This nonspecificity results in numerous adverse effects—in particular, myelosuppression, gastrointestinal toxicity, and damage to normal tissues leading to significant morbidity. Although chemotherapy can be an effective component in palliative care or in certain combination settings, its overall contribution as a monotherapy in melanoma is limited when compared to the dramatic responses seen with immunotherapy or targeted therapy.

Comparative Analysis and Effectiveness
Efficacy of Different Drug Classes
A comparative view of the drug classes in melanoma treatment reveals distinct differences in efficacy.
Immunotherapy has emerged as a transformative approach; in numerous clinical trials, immune checkpoint inhibitors have shown the potential to deliver durable responses and long-term survival benefits in patients with advanced melanoma. In selected patient populations, especially those who exhibit robust immune-mediated tumor rejection, responses can be both deep and lasting. However, it is important to note that a significant subset of patients either do not respond or eventually relapse, underscoring the underlying heterogeneity of melanoma and the need for biomarkers and combination strategies.

Targeted therapy, particularly for patients with BRAF mutations, results in rapid tumor shrinkage and significant improvements in progression-free survival. Combination regimens of BRAF and MEK inhibitors are now standard and have resulted in higher response rates compared with monotherapy. The response with targeted therapy tends to be faster; however, the durability is challenged by the emergence of resistance mechanisms over time. As such, although targeted therapies provide impressive initial responses, many patients eventually experience disease progression.

Chemotherapy remains the least effective of the three classes in terms of objective response rates and overall survival. With response rates in advanced melanoma generally in the single digits, chemotherapy is now reserved primarily for patients who have exhausted other treatment options or for cases in which other modalities are contraindicated. The modest benefits of chemotherapy contrast sharply with the substantial, albeit variable, success seen with the more modern treatments.

Side Effects and Safety Profiles
Each drug class comes with a unique set of adverse effects that impact tolerability and patient quality of life.

Immunotherapy is notable for immune-related adverse events (irAEs), which are a consequence of an overactivated immune system. These adverse events can affect any organ system but are most commonly observed in the skin, gastrointestinal tract, liver, endocrine organs, and lungs. While many irAEs are manageable with prompt recognition and immunosuppressive treatments such as corticosteroids, some can be severe or even life threatening. The balance between eliciting a powerful antitumor immune response and moderating systemic toxicity represents a key challenge in clinical practice.

Targeted therapy generally exhibits a more predictable toxicity profile. BRAF inhibitors, for instance, are associated with dermatologic toxicities including rash, photosensitivity, and the paradoxical formation of cutaneous squamous cell carcinomas, while MEK inhibitors may cause peripheral edema, rash, and gastrointestinal symptoms. The combination of BRAF and MEK inhibitors has been shown to minimize some of the cutaneous effects while still presenting risks such as fever, fatigue, and cardiotoxicity. Importantly, the adverse event profile is usually less immunologically mediated but the overall tolerability can be limited by the rapid emergence of on-target toxicities.

Chemotherapy is accompanied by a broad array of side effects because of its non-specific mechanism of action. Notable toxicities include myelosuppression, gastrointestinal disturbances (nausea, vomiting, diarrhea), alopecia, and potential long-term organ damage. Given its systemic cytotoxicity, chemotherapy is generally less well tolerated over prolonged periods compared to the more targeted approaches.

Combination Therapies
One of the most promising responses to the limitations of individual drug classes in melanoma treatment has been the development of combination therapies. These combinations may take the form of dual immunotherapy, the combination of targeted therapies (such as BRAF with MEK inhibitors), or even cross-class combinations that integrate immunotherapy with targeted agents.

Dual checkpoint blockade—for example, combining anti–CTLA-4 with anti–PD-1 therapies—has demonstrated improved overall survival and higher response rates compared to monotherapy. However, this comes at the cost of increased toxicity, which necessitates careful patient selection and management strategies to mitigate severe irAEs.

Similarly, the combination of BRAF and MEK inhibitors represents a paradigm shift in targeted therapy. By simultaneously obstructing sequential kinases in the MAPK pathway, these regimens not only enhance tumor response rates but also delay the onset of resistance that is so frequently encountered with single-agent therapy. The rationale for such combinations is supported by preclinical and clinical evidence that the pathways driving tumor survival are interconnected; hence, dual inhibition can produce synergistic antitumor effects.

There are also emerging strategies that combine agents from different classes. For example, the combination of immunotherapy with targeted therapy is an area of intense research. The idea is that targeted agents can rapidly debulk tumors and modulate the tumor microenvironment (e.g., by decreasing immunosuppressive cytokines or increasing antigen presentation), thus rendering residual tumor cells more susceptible to immune-mediated attack. Preliminary clinical studies suggest that such combinations could improve outcomes over either strategy alone, but these approaches require optimized scheduling and dosing to balance efficacy and toxicity.

Conclusion
In summary, melanoma treatment has evolved dramatically, shifting from the historical reliance on chemotherapy and surgery to a more nuanced, biomarker-driven approach utilizing immunotherapy and targeted therapy.

An overview of melanoma reveals that this aggressive cancer originates from melanocytes, has a rising incidence due to environmental and genetic factors, and is associated with high mortality in its advanced stages. The current treatment landscape now includes multiple systemic therapies that have redefined how clinicians manage advanced disease, with immunotherapy and targeted therapy playing central roles alongside traditional chemotherapy.

Different drug classes used in treating melanoma work through diverse biological mechanisms. Immunotherapy works by modulating the immune system—especially T-cell–mediated responses—through the blockade of inhibitory checkpoints such as CTLA-4 and PD-1, effectively restoring an impaired antitumor immune response. In contrast, targeted therapy is designed to specifically interrupt dysregulated signaling pathways, such as the BRAF–MAPK cascade, that drive melanoma cell proliferation and resistance to apoptosis; combination targeted therapies (BRAF plus MEK inhibitors) have become the standard for BRAF-mutated melanoma owing to their synergistic effects and improved response rates. Chemotherapy, on the other hand, employs cytotoxic agents that compromise DNA replication and induce apoptosis in rapidly dividing cells, but its efficacy is often blunted by intrinsic chemoresistance and widespread toxicity.

Comparatively, immunotherapy has been transformational in terms of durability and long-term survival benefits despite its risk of immune-related adverse events, while targeted therapy provides rapid responses but is limited by the eventual emergence of resistance. Chemotherapy remains a fallback option with lower efficacy and higher systemic toxicity. Rational combination therapies—whether combining dual immunotherapeutics, pairing targeted agents, or integrating across classes—are increasingly being adopted to harness synergistic benefits and overcome individual limitations. The advent of combinations, such as dual checkpoint blockade or triple therapy regimens integrating targeted agents with immunotherapy, exemplify the strategic evolution in melanoma management, aiming to improve overall survival while balancing safety profiles.

The future of melanoma treatment lies in a tailored, multidisciplinary approach that incorporates genomic profiling, real‐time immune monitoring, and a combination of drug classes to both potentiate tumor regression and delay or overcome resistance mechanisms. While challenges remain—including managing toxicities and optimal sequencing of therapies—the advancements in understanding the molecular and immunologic underpinnings of melanoma have paved the way for promising new therapeutic strategies that can ultimately offer improved outcomes and quality of life for patients with this aggressive malignancy.

In conclusion, the integration of diverse drug classes—immunotherapy, targeted therapy, and chemotherapy—not only represents an evolution in our scientific understanding of melanoma but also marks a practical shift toward more effective and personalized treatment regimens. The general strategy is to initiate potent tumor debulking (especially with targeted agents when applicable), awaken and sustain the immune response against the tumor (via immunotherapy), and, if needed, use chemotherapy as an adjunct to manage resistant disease or to palliate symptoms. From multiple perspectives—molecular, clinical, immunologic, and economic—the current paradigm emphasizes the necessity of combination therapies, optimized sequencing, and patient-specific tailoring to achieve the best outcomes in melanoma treatment.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.