What are the new drugs for Prostate Cancer?

17 March 2025

Overview of Prostate Cancer

Prostate cancer remains one of the most commonly diagnosed malignancies in men worldwide and continues to be a leading cause of cancer‐related mortality. Over the last several decades, early detection has improved with widespread use of screening tools such as prostate‐specific antigen (PSA) tests and digital rectal exams. These screening approaches have led to a stage migration where many prostate cancers are detected early. However, a significant proportion of patients present with advanced disease, and even when localized cancers are detected, there is substantial heterogeneity in tumor biology and clinical course. Recent epidemiological studies indicate that while the incidence of prostate cancer is high, particularly in Western societies, mortality in localized cases remains relatively low compared to those developing metastatic castration‐resistant prostate cancer (mCRPC). In addition, improved awareness of risk factors such as ethnicity, family history, and genetic mutations has contributed to evolving clinical management strategies, emphasizing the need for personalized treatment decisions based on molecular and clinical characteristics.

Risk Factors and Diagnosis

Risk factors for prostate cancer include non‐modifiable parameters such as increasing age, family history of prostate or related cancers (for example, breast cancer) and race/ancestry, as well as modifiable factors such as diet, body weight, and physical activity. Research examining the underlying genetic determinants has pointed out that alterations in key genes such as BRCA1, BRCA2, and other DNA repair genes contribute to a higher disease risk and often to more aggressive forms of the disease. Diagnosis is typically initiated with PSA screening, which is sensitive and widely used though not perfectly specific, often followed by imaging techniques and ultimately biopsy confirmation. Recently, advanced imaging agents such as radiolabeled prostate-specific membrane antigen (PSMA) tracers have improved the detection of metastases, thereby assisting clinicians in better staging the disease.

Recent Advancements in Prostate Cancer Treatment

Newly Approved Drugs

Over the past decade, the therapeutic landscape for prostate cancer has witnessed a significant transformation. One of the most notable changes has been the introduction of several new drugs that are designed to improve overall survival, delay progression, and offer treatment alternatives even when initial hormone-based therapies fail. The newly approved drugs now primarily target either the androgen receptor (AR) signaling axis or other specific molecular pathways that drive cancer cell survival.

A major breakthrough came with the approval of androgen deprivation therapies that specifically target the AR pathway. Abiraterone acetate (marketed as Zytiga) and enzalutamide (marketed as Xtandi) revolutionized the treatment of mCRPC by inhibiting androgen production and AR signaling respectively. Abiraterone works by inhibiting the enzyme CYP17A1, an essential catalyst in androgen biosynthesis, while enzalutamide directly blocks the receptor, both leading to significant clinical benefit by delaying disease progression and extending survival. Subsequent to these approvals, other agents have been approved based on the data from large-scale randomized clinical trials.

Cabazitaxel (marketed as Jevtana) is a taxane chemotherapy agent that was approved for patients who have progressed on docetaxel therapy. It is specifically designed for mCRPC and has been shown to overcome resistance to previous taxane treatment, providing an important second-line therapy option. Radium-223 chloride (marketed as Xofigo) represents another class of therapeutic agents: targeted alpha therapy. This radiopharmaceutical selectively targets bone metastases—a frequent clinical problem in advanced prostate cancer—and has been approved on the basis that it significantly prolongs overall survival while improving the quality of life.

In addition to the agents mentioned above, there has been growing regulatory interest in poly (ADP-ribose) polymerase (PARP) inhibitors for patients with DNA repair gene mutations. Rucaparib and olaparib are two PARP inhibitors that have recently been approved for subpopulations of patients with mCRPC harboring alterations in BRCA1/2 or other homologous recombination repair genes. These drugs exploit the concept of synthetic lethality, providing clinical benefit in patients whose tumors have impaired DNA repair pathways. Many of these drugs have undergone rigorous evaluation in advanced phase clinical trials, demonstrating not only efficacy but also manageable safety profiles.

The newly approved drugs have been welcomed by the clinical community for their potential to expand therapeutic options. Their approvals are based on data from multicenter phase III trials that assess clinically meaningful endpoints such as overall survival, progression‐free survival, and quality-of-life improvements. These drugs have set a new standard of care and are currently being integrated into treatment algorithms in diverse clinical scenarios—from hormone-sensitive to castration-resistant disease. In the synapse reference collection, several sources confirm the clinical impact of these agents and underscore their role as new standards in prostate cancer management.

Drugs in Clinical Trials

In parallel with the approval of several new agents, a wide range of drugs is currently under investigation. These drugs represent not only extensions of the mechanisms already targeted by approved agents but also novel approaches based on emerging understanding of prostate cancer biology.

Among the promising drugs in clinical trials are newer generation anti-androgens that may prove effective even in cases resistant to current hormonal therapies. Novel androgen receptor degraders and next-generation androgen receptor antagonists are being evaluated for their ability to more completely shut down signaling, especially in tumors with AR splice variants that are not blocked by conventional agents. Some early phase trials have reported that improved receptor-targeted approaches may offer a potent means of delaying or reversing castration resistance.

Furthermore, numerous targeted therapies outside of the hormonal axis are in early-phase clinical evaluation. For example, agents targeting the phosphatidylinositol 3-kinase (PI3K)–AKT–mTOR pathway, which has been implicated in therapy resistance, are under study. Drugs that target this pathway may be particularly effective when used in combination with AR inhibitors, given the known cross-talk between these pathways. In addition, there is significant interest in combining targeted therapies with established treatments to overcome resistance mechanisms.

Immunotherapy is another frontier in prostate cancer treatment. Although prostate cancer is generally considered an “immunologically cold” tumor, recent clinical trials are investigating the potential of checkpoint inhibitors either alone or in combination with other treatments such as vaccines or other immunomodulatory drugs. Early signals from trials combining PD-1/PD-L1 inhibitors with anti-androgen therapies or radiopharmaceuticals have been encouraging, although further studies are needed to delineate which patients are most likely to benefit.

Several antibody-drug conjugates (ADCs) and bispecific T-cell engagers (BiTEs) are also in clinical trials for prostate cancer. These innovative agents aim to combine the specificity of targeted antibodies with the cytotoxic potency of chemotherapeutics or immune cell activation. By harnessing the specificity of antibodies against tumor-associated antigens, these conjugates promise to improve the therapeutic index and minimize systemic toxicity. Although most of these agents are in early clinical development, the preliminary data appear promising and may offer new treatment avenues in the near future.

Other investigational agents include novel radiopharmaceuticals beyond radium-223 that target specific molecular markers expressed on prostate cancer cells, such as PSMA. The development of new PSMA-targeted radionuclide therapies is aimed at delivering potent radiation doses directly to cancer cells, thereby reducing systemic side effects. These agents are now entering phase II and III studies and may soon complement or even supersede existing treatments.

Mechanisms of Action

Targeted Therapies

The mechanism of action of the new drugs for prostate cancer largely centers on improved targeting of specific molecular pathways that drive tumor growth and resistance. Targeted therapies such as abiraterone acetate and enzalutamide inhibit androgen synthesis and receptor activation directly, thus depriving prostate cancer cells of the growth stimulus provided by androgens. This is a critical advance because, in many cases, prostate cancer remains dependent on the androgen receptor (AR) signaling axis even after development of castration resistance.

In addition to AR-targeted agents, PARP inhibitors such as rucaparib and olaparib work by interrupting the DNA damage repair mechanisms that cancer cells rely on when one pathway is compromised by mutations. By blocking PARP, these drugs increase DNA damage in cells harboring defective homologous recombination repair, ultimately leading to cell death via synthetic lethality. This mechanism has proven highly beneficial in patient subpopulations with genetic alterations in DNA repair genes.

Another promising approach involves targeting intracellular signaling pathways that contribute to resistance. Agents under investigation aim to inhibit the PI3K–AKT–mTOR pathway, which is frequently activated in prostate cancer and can compensate for inhibited AR signaling. By combining AR pathway inhibitors with inhibitors of this alternative survival pathway, researchers hope to overcome resistance mechanisms that limit the efficacy of current treatments.

Emerging treatments in early-phase trials also include antibody-drug conjugates (ADCs) that couple a highly specific monoclonal antibody—targeting surface antigens overexpressed on prostate cancer cells—with a potent cytotoxic drug. This design ensures that the cytotoxic agent is delivered selectively to cancer cells while sparing normal tissues. Similarly, bispecific T-cell engagers (BiTEs) are designed to redirect cytotoxic T cells to tumor cells, thereby eliciting an immune response directly against the cancer. These strategies represent an innovative approach to targeted therapy, combining precise molecular targeting with robust cell-killing mechanisms.

Hormonal Therapies

Hormonal therapies remain cornerstone treatments for prostate cancer due to the tumor’s reliance on androgen signaling. Traditional androgen deprivation therapy (ADT) through surgical castration or luteinizing hormone-releasing hormone (LHRH) agonists has been a standard of care for decades. However, the development of resistance has driven the need for new hormonal therapies that can effectively target the androgen receptor even in low-androgen environments.

The new generation of hormonal therapies focuses on both inhibiting androgen production and blocking AR activity. Abiraterone acetate, for example, acts as a CYP17A1 inhibitor, dramatically reducing the synthesis of androgens in the adrenal glands and within tumor tissues. Enzalutamide, on the other hand, is a potent AR antagonist that not only prevents androgen binding but also interferes with AR nuclear translocation and DNA binding, thereby effectively shutting down receptor signaling even in castration-resistant disease.

Research into overcoming resistance has led to novel drugs that degrade the androgen receptor protein, including compounds known as AR degraders. These agents are being developed to specifically target AR splice variants that are commonly expressed in mCRPC and are not effectively inhibited by conventional anti-androgens. Such therapies hold promise for patients who no longer respond to drugs like abiraterone or enzalutamide due to the presence of these splice variants.

Overall, hormonal therapies have evolved from simply reducing systemic androgen levels to a sophisticated, multi-pronged approach that includes enzyme inhibitors, receptor blockers, and agents targeting mutant or splice variant receptors. This evolution reflects a more detailed understanding of the interplay between hormonal signaling and prostate cancer biology.

Clinical Efficacy and Safety

Clinical Trial Results

The clinical trial evidence supporting the efficacy of the new drugs for prostate cancer has provided compelling data on both survival benefits and quality-of-life improvements. Large phase III trials of abiraterone acetate and enzalutamide have demonstrated significant improvements in overall survival, progression-free survival, and other outcome measures compared to placebo or previous standard treatments. For instance, trials have shown that abiraterone and enzalutamide can delay disease progression by several months and in some cases improve overall survival by more than 3–5 months, which is clinically meaningful in the context of advanced disease.

Cabazitaxel, another newly approved agent, has also been evaluated in randomized controlled trials where it significantly improved overall survival in patients who had progressed on docetaxel, illustrating its role as a critical second-line therapy. Radium-223 chloride trials demonstrated not only survival benefits but also improvements in pain control and quality of life for patients with bone metastases. In patients with DNA repair gene mutations, PARP inhibitors such as rucaparib and olaparib have shown high response rates and durable disease control in early phase studies, leading to their approval for specific patient subgroups.

Early-phase clinical trials of investigational agents, including novel AR degraders, PI3K–AKT–mTOR inhibitors, and promising immunotherapies, have yielded encouraging preliminary results. These studies often incorporate biomarker-driven patient selection to ensure that the therapeutic effect is maximized in the appropriate subpopulations. The encouraging safety profiles seen in many of these trials have paved the way for subsequent trials while reinforcing the potential for these drugs to be added to the arsenal against prostate cancer.

Side Effects and Management

Safety profiles and side effects are critical factors when considering new treatments for prostate cancer. Although the new drugs generally offer improved efficacy over traditional therapies, they are not without adverse effects. The side effects associated with abiraterone include hypertension, hypokalemia, and liver function abnormalities; these are generally managed with corticosteroids and supportive care measures. Enzalutamide is associated with adverse events such as fatigue, hypertension, and rarely, seizures; nonetheless, its overall tolerability has been acceptable.

Cabazitaxel, as a chemotherapeutic agent, does present a risk of neutropenia, febrile neutropenia, and gastrointestinal toxicity; however, careful dose management and supportive interventions such as granulocyte colony-stimulating factors (G-CSF) have allowed it to be safely administered to the appropriate patient population. Radium-223’s toxicity profile largely centers on hematologic adverse events such as anemia and thrombocytopenia but with careful patient selection and monitoring these side

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.