How do different drug classes work in treating Metastatic castration-resistant prostate cancer?

17 March 2025

Overview of Metastatic Castration-Resistant Prostate Cancer (mCRPC)

Metastatic castration-resistant prostate cancer is defined as prostate cancer that has continued to progress despite achieving castrate levels of testosterone through androgen deprivation therapy (ADT). Initially, patients respond well to ADT; however, over time cancer cells adapt by reactivating androgen receptor (AR) signaling or activating alternative survival pathways. This leads to a phenotype that is no longer hormone-sensitive and is characterized by continued tumor growth and metastasis even in a low-androgen environment. In addition, the tumor develops molecular aberrations such as AR amplification, mutations, and the emergence of splice variants, which allow prostate cancer cells to obtain intracellular androgens or remain responsive to minimal hormonal stimuli. In a broader sense, mCRPC reflects a disease state where multiple resistance mechanisms, including changes in DNA repair genes or activation of alternative proliferative pathways, contribute to its aggressiveness and eventual fatal outcome. These cellular and molecular changes set the stage for therapeutic interventions that are designed to target not only the androgen signaling axis but also other survival mechanisms.

Current Treatment Landscape 
The treatment landscape for mCRPC has evolved significantly over the past two decades. Early treatments were based solely on hormonal manipulation – primarily reducing circulating testosterone. As mCRPC emerged, treatment strategies expanded to include newer generation hormonal therapies, chemotherapeutic agents such as docetaxel and cabazitaxel, and more recently, immunotherapy and targeted agents like PARP inhibitors (niraparib, olaparib, and rucaparib) for selected patients. In clinical practice, leveraging combinations of these drug classes in sequential or even concurrent strategies is becoming common, with the goal of harnessing complementary mechanisms of action that may overcome resistance and prolong survival. In some instances, bispecific antibodies and combination regimens with immune checkpoint inhibitors have also been explored to re-engage the immune system against prostate cancer cells. The overall approach is becoming one of precision medicine, whereby treatment decisions are informed by molecular profiling and clinical markers, aiming to maximize efficacy while managing toxicity.

Drug Classes Used in mCRPC Treatment

Hormonal Therapies 
Hormonal therapies continue to form the foundation for mCRPC treatment, albeit through more advanced agents. These drugs target the androgen receptor (AR) pathway directly or indirectly and include agents such as abiraterone acetate, enzalutamide, apalutamide, and darolutamide. Abiraterone acetate works by inhibiting cytochrome P450 17A1 (CYP17A1) enzyme activity, thereby reducing both adrenal and intratumoral androgen synthesis. Enzalutamide and similar agents act directly as AR antagonists—they bind with high affinity to the AR ligand-binding domain, blocking the receptor’s activation, nuclear translocation, and interaction with androgen-responsive genes. Over time, as prostate cancer cells adapt to hormone suppression, these agents continue to be useful even in low-androgen environments, though resistance mechanisms (such as AR splice variants) may limit their long-term effectiveness. Hormonal therapies are usually well tolerated relative to chemotherapy, which makes them attractive as first-line agents and in combination with other modalities for patients with mCRPC.

Chemotherapy Agents 
Chemotherapy remains a vital option for patients with mCRPC, particularly after progression on hormonal agents. Docetaxel was the first chemotherapeutic agent to show a survival benefit for mCRPC patients, leading to its adoption as a standard of care in the early 2000s. Docetaxel works by disrupting microtubule function in dividing cells, which leads to cell-cycle arrest and subsequent apoptosis of rapidly dividing cancer cells. Following docetaxel, cabazitaxel emerged as a second-line chemotherapy; it was specifically designed to overcome resistance to docetaxel. Both agents are usually administered with corticosteroids (such as prednisone) to mitigate inflammation and to improve tolerability while managing side effects. While chemotherapy can offer significant tumor shrinkage and delay disease progression, its use is associated with systemic toxicities, particularly hematologic toxicity, which necessitates careful dosing and patient selection.

Immunotherapy Options 
The advent of immunotherapy in mCRPC is more recent compared to hormonal and chemotherapeutic strategies. Although mCRPC is typically considered a “cold” tumor regarding immune infiltration, increasing efforts have been made to engage antitumor immunity via various mechanisms. Immune checkpoint inhibitors (ICIs) that target proteins such as PD-1 and PD-L1 aim to reverse immune suppression within the tumor microenvironment and restore effective cytotoxic T-cell responses. In addition, emerging approaches include bispecific antibodies that simultaneously target tumor-specific antigens (for example, prostate-specific membrane antigen, PSMA) and immune effectors like CD3 or costimulatory receptors (e.g., CD28), thereby bridging T cells with cancer cells to induce targeted cell killing. Other investigational immunotherapies include vaccine-based approaches and combination regimens of ICIs with hormonal therapies, which may have synergistic effects by modulating the immune response and reducing tumor-induced immunosuppression. Although immune-based therapies have shown promising activity in other malignancies, their use as monotherapy in mCRPC has been challenging, and ongoing investigations aim to better define their role in combination treatment strategies.

Mechanisms of Action

Mechanisms of Hormonal Therapies 
Hormonal therapies primarily work by disrupting the androgen signaling axis, which is central to the growth and survival of prostate cancer cells. 
• Abiraterone acetate inhibits CYP17A1, an enzyme pivotal in androgen biosynthesis. By blocking both the 17α-hydroxylase and 17,20-lyase activities, abiraterone reduces the conversion of precursors into active androgens, thus depleting the androgens available both systemically and within the tumor microenvironment. 
• Direct AR antagonists—such as enzalutamide, apalutamide, and darolutamide—bind to the androgen receptor, preventing it from undergoing the conformational changes necessary for activation. As a consequence, receptor dimerization, nuclear translocation, and binding to androgen response elements on DNA are inhibited, effectively suppressing androgen-regulated gene transcription. This blockade impairs cell cycle progression and induces apoptosis, even in resistant tumor cell subpopulations that express mutant or overexpressed AR proteins. Moreover, these drugs have been engineered to bind with higher affinity than earlier generations of antiandrogens, thereby addressing resistance mediated by receptor overexpression. 
• Furthermore, some novel approaches using bispecific antibodies—designed to simultaneously bind PSMA on tumor cells and CD28 or CD3 on T cells—provide an indirect hormonal approach by not only targeting an antigen specific to prostate cancer but also activating immune effector functions. Although these are primarily categorized under immunotherapy, they have a direct impact on androgen-driven cancers by localizing the immune response to prostate cancer cells.

Mechanisms of Chemotherapy Agents 
Chemotherapy agents for mCRPC operate mainly through cytotoxic effects that disrupt cell division and survival: 
• Docetaxel and cabazitaxel belong to the taxane class, and their mechanism involves the stabilization of microtubules. This stabilization prevents the dynamic reorganization of the microtubule network that is necessary for mitotic spindle formation, thereby halting the cell cycle at the G2/M phase and inducing apoptosis due to prolonged mitotic arrest. 
• In addition to direct microtubule stabilization, docetaxel has been reported to induce immunogenic cell death in cancer cells, which might also contribute to secondary immune activation through the release of danger signals and tumor antigens. This phenomenon creates a potential synergy between chemotherapy and immunotherapy, as dying cancer cells can stimulate antigen-presenting cells and promote subsequent T-cell mediated responses. 
• Chemotherapy has also been observed to work in conjunction with corticosteroids (for example, prednisone) to reduce inflammation and manage adverse effects, thus indirectly improving drug tolerability. However, the systemic cytotoxicity can manifest as hematologic toxicity, neuropathy, and gastrointestinal adverse events, which require careful management and often limit the doses that can be administered.

Mechanisms of Immunotherapy 
Immunotherapy in mCRPC is an evolving field aimed at reactivating the patient’s own immune system to target prostate cancer cells. 
• Immune checkpoint inhibitors (ICIs) act by blocking inhibitory signals that dampen T-cell activity. In mCRPC, blockade of PD-1 on T cells or PD-L1 expressed on tumor cells can lead to reactivation of T-cell function, restoring the ability of T cells to recognize and attack cancer cells. Despite prostate cancer’s characterization as an immunologically “cold” tumor, the underlying immune reactivity can be augmented, especially when combined with other therapies that increase antigen presentation or decrease immunosuppressive factors. 
• Bispecific antibodies represent another innovative immunotherapeutic approach. By linking T cells directly with cancer cells—through one arm that binds to a tumor-associated antigen (such as PSMA) and another arm that engages CD3 or costimulatory receptors (e.g., CD28)—these drugs facilitate the formation of an immunological synapse. This forced proximity can trigger potent, localized cytotoxic responses against cancer cells, bypassing some of the limitations associated with inadequate T-cell infiltration in the tumor. 
• Additional immunotherapy strategies include therapeutic vaccines that prime the immune system against prostate-specific antigens and adoptive cell therapies, which involve the ex vivo expansion and reinfusion of tumor-infiltrating lymphocytes or genetically modified T cells. The idea behind these modalities is to generate a targeted immune response that can overcome the immunosuppressive tumor microenvironment (TME), though these approaches are still under active investigation for mCRPC.

Clinical Effectiveness and Considerations

Efficacy and Survival Benefits 
Clinical trials and real-world studies have demonstrated that each drug class offers distinct benefits in mCRPC: 
• Hormonal therapies have significantly improved overall survival when compared to historical standard ADT alone. Agents such as abiraterone and enzalutamide have been associated with prolonged progression-free survival (PFS) and overall survival (OS) in large phase III trials. Their mechanism of directly targeting the androgen receptor pathway ensures that even hormone-independent tumor populations maintain some dependence on residual AR signaling, which can be effectively suppressed by these agents. 
• Chemotherapeutic agents, particularly docetaxel, provided the first evidence of a survival benefit in mCRPC. Docetaxel-based regimens have shown median survival benefits in the range of several months over previous non-cytotoxic therapies, and cabazitaxel has further improved outcomes in patients who progress after docetaxel treatment. Moreover, there is evidence that taxane treatment not only reduces tumor burden through cytotoxic effects but may also trigger an immunogenic response, potentially contributing to longer-term benefits when used in combination with other agents. 
• Immunotherapy has shown promising response rates in other tumor types; however, as monotherapy in mCRPC, its results have been modest. Nonetheless, when used in combination with hormonal therapies, chemotherapy, or bispecific antibody approaches, immunotherapy can improve overall survival and provide durable responses in selected patients. The synergistic effects observed in combination regimens suggest that immunotherapy may be most effective when used as part of a multi-agent strategy that includes other modalities to modulate the tumor microenvironment and enhance immune recognition.

Side Effects and Management 
Each therapeutic class has its own safety profile and potential side effects that must be managed carefully: 
• Hormonal therapies are generally well tolerated relative to chemotherapy. Common side effects include fatigue, hot flashes, and hypertension; however, these are typically less severe than the cytotoxic side effects seen with chemotherapy. Long-term hormonal suppression, however, may lead to metabolic alterations and bone density loss, requiring supportive care measures such as bisphosphonates or denosumab. 
• Chemotherapy agents like docetaxel and cabazitaxel are associated with a higher incidence of side effects, including neutropenia, neuropathy, fatigue, and gastrointestinal disturbances. The use of corticosteroids as adjuncts helps to mitigate some of these side effects, but support with growth factor support and dose modifications are often necessary to keep treatment tolerable, especially in older or frail patients. 
• Immunotherapy can trigger immune-related adverse events (irAEs) such as colitis, dermatitis, pneumonitis, and endocrinopathies due to the non-specific reactivation of the immune system. Close monitoring, early detection, and prompt intervention with immunosuppressive therapies (for example, corticosteroids) are essential for managing these risks. The combination of drug classes, particularly when immune activation is paired with cytotoxic agents, requires careful scheduling and dosing to minimize overlapping toxicities while retaining efficacy.

Patient Selection Criteria 
Patient selection in mCRPC is a multifactorial process driven by tumor biology, previous treatment history, patient performance status, and molecular markers: 
• Hormonal therapies are often used as a first-line or early second-line treatment in patients who maintain adequate performance status and whose tumors show evidence of AR-driven proliferation. Patients with detectable AR splice variants or mutations may require newer generation AR antagonists that have higher binding affinities or broader antagonistic effects. 
• Chemotherapy is typically reserved for patients who have progressed on hormonal therapies, often those with rapidly progressive disease as indicated by a high tumor burden or symptomatic metastases. Since chemotherapy is associated with higher toxicity, patient age, comorbidities, and bone marrow reserve are critical considerations. 
• Immunotherapy or combination immunotherapeutic approaches are considered for patients with indications of an active immune response, such as those with increased PD-L1 expression or concurrent markers of genomic instability (for example, defects in DNA repair machinery). Given that mCRPC is generally immunologically “cold,” such approaches are best reserved for patients in whom combination strategies (often including hormonal therapy or chemotherapy) are feasible and where tumor profiling may demonstrate potential neoantigens. 
• Furthermore, emerging biomarkers and genomic profiling techniques are being incorporated into clinical practice to help predict responses and identify patients who may benefit more from one therapeutic strategy over others. Molecular markers such as AR gene amplification, DDR (DNA damage repair) gene mutations, and even emerging immune signatures are increasingly used to personalize treatment regimens and monitor safety.

Conclusion 
In summary, the treatment of metastatic castration-resistant prostate cancer employs an integrated multi-modality approach that relies on distinct drug classes with complementary mechanisms of action. Hormonal therapies continue to target residual androgen signaling and have evolved to overcome resistance with agents such as abiraterone and enzalutamide. Chemotherapy, primarily through taxane agents like docetaxel and cabazitaxel, provides cytotoxicity by disrupting critical cellular structures and may also induce immunogenic cell death. Immunotherapy, though still challenging in mCRPC due to the “cold” tumor microenvironment, is gaining traction through combination strategies such as bispecific antibodies that bring immune cells into close apposition with tumor cells. 

From a general perspective, each drug class contributes unique benefits to the overall survival and disease control. The specific mechanisms are driven by targeted inhibition of androgen synthesis and receptor activation, cytotoxic disruption of cell division, and immune activation through checkpoint blockade or cell bridging. Looking at these mechanisms from a broader perspective, the integration of these drug classes is a classic example of precision medicine in oncology, where a detailed understanding of tumor biology drives the selection, sequencing, and combination of therapies.

From a specific perspective, hormonal therapies work by both depleting the ligands required for receptor activation and directly antagonizing the receptor itself, thereby crippling a central survival pathway for prostate cancer cells. Chemotherapy agents, depending on their molecular targets, employ microtubule stabilization and cell cycle disruption to induce apoptosis in rapidly dividing cells. Immunotherapeutic approaches, although in an evolving phase, attempt to shift the balance within the tumor microenvironment—either by directly reinvigorating T-cell responses via checkpoint inhibitors or by fostering a cytotoxic response through bispecific antibody engagement. Each approach is optimized for different clinical scenarios based on the patient’s performance status, prior treatment history, and the molecular characteristics of the tumor.

From a general-specific-general standpoint, the overarching strategy in mCRPC management is to overcome therapeutic resistance by attacking the cancer on several fronts simultaneously. The general goal is to prolong survival and improve quality of life for patients with an otherwise lethal disease. Specifically, through the development and deployment of advanced hormonal therapies and cytotoxic agents—with a growing complement of immunotherapeutic strategies—clinicians are now able to personalize treatment and adjust therapeutic regimens in response to evolving tumor biology. This paradigm shift from a single-agent approach to a combination strategy not only improves the chances of response but also addresses the multifactorial mechanisms of resistance inherent in mCRPC.

In conclusion, understanding how different drug classes work in treating mCRPC is critical for the development of rational combination therapies. Hormonal therapies interrupt androgen signaling, chemotherapy agents induce direct cytotoxicity and in some cases stimulate immunogenic cell death, and immunotherapies aim to reverse immune suppression in the tumor microenvironment. Together, these approaches offer a multi-pronged strategy that has transformed the mCRPC treatment landscape over the past two decades. Ongoing investigations, biomarker-driven patient selection, and combination strategies promise further improvements in clinical outcomes for patients facing this challenging disease. The future of mCRPC treatment lies in the synergistic use of these drug classes to achieve longer survival, better quality of life, and ultimately, a more hopeful outlook for patients with advanced prostate cancer.

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.