Combination Treatments for Advanced Prostate Cancer (2025)

Advanced prostate cancer refers to disease that has spread beyond the prostate gland or has become resistant to initial treatment. In recent years, studies have shown that using more than one therapy at the same time (“combination therapy”) can help men with advanced prostate cancer live longer and feel better​. In fact, “one drug alone is no longer sufficient” – combining therapies improves survival without worsening quality of life in most patients​. This guide covers current approved treatments, a comparison of effectiveness and side effects, prognosis and survival rates, emerging therapies in trials, managing side effects, and tips for talking with doctors. The information is based on high-impact clinical studies from the last seven years, explained in clear language.

 

1. Approved Treatments for Advanced Prostate Cancer

Advanced prostate cancer is usually treated with a combination of therapies. Below are the key FDA-approved and internationally accepted treatments, each with an explanation of how they work (mechanism), how they are given (administration), and their clinical benefits (outcomes). All of these treatments have strong evidence from clinical trials.

Androgen Deprivation Therapy (ADT) – Foundation of Treatment

Mechanism: Prostate cancer typically depends on androgens (male hormones like testosterone) to grow. ADT reduces the body’s production of testosterone (either by medication or surgery). Medications like leuprolide or goserelin are injections that signal the testes to stop making testosterone, while drugs like degarelix block the signal altogether​. Surgical removal of the testes (orchiectomy) also eliminates most testosterone production.
Administration: ADT by injection is given on a regular schedule (monthly or every 3–6 months), or orchiectomy is a one-time outpatient surgery.
Outcomes: ADT is very effective at initially shrinking advanced tumors and lowering PSA levels. Almost all patients respond initially, experiencing cancer regression and symptom relief. However, the cancer usually adapts over time and can progress despite low hormone levels (becoming castration-resistant). ADT alone was the standard of care for decades, but ADT by itself is no longer considered enough for most metastatic cases​. It is now used in combination with other therapies to improve survival. Common side effects: ADT causes low testosterone, leading to hot flashes, reduced sexual function, fatigue, muscle loss, weight gain, and bone thinning over time. These side effects are often managed with exercise, diet, calcium/Vitamin D, and medications for bone health or hot flashes.

 

Androgen Receptor Pathway Inhibitors (ARPIs) – Advanced Hormonal Therapies

Mechanism: Even after testosterone from the testes is reduced by ADT, prostate cancer cells can still find ways to feed on androgens (from the adrenal glands or within the tumor). AR pathway inhibitors block the cancer’s ability to use any remaining hormones​. There are two main types:

 

  • Androgen synthesis inhibitor: Abiraterone (Zytiga) is a pill that blocks an enzyme (CYP17) needed to make androgens. By stopping all internal production of testosterone, it “starves” the cancer of hormones. It’s given with low-dose prednisone to prevent side effects from hormone imbalance.
  • Androgen receptor blockers: Enzalutamide (Xtandi), Apalutamide (Erleada), and Darolutamide (Nubeqa)are oral medications that attach to the androgen receptor in cancer cells, so the hormone can’t activate it. Essentially, they prevent cancer cells from using testosterone that may still be in the body​. These drugs are more potent than older drugs like bicalutamide.Administration: All are taken as daily pills. Abiraterone is taken on an empty stomach with prednisone pills; enzalutamide, apalutamide, or darolutamide are taken daily (darolutamide is twice daily) and do not require steroid use.
    Outcomes: Adding any of these ARPIs to ADT has dramatically improved patient outcomes. In men with metastatic hormone-sensitive prostate cancer (mHSPC), ADT + abiraterone lowered the risk of death by ~38% in the LATITUDE trial​ onclive.com. After about 3 years follow-up, 66% of patients on abiraterone were alive vs. 49% on ADT alone​. Median survival on ADT alone was about 34.7 months, while it was not reached (over 3 years) with abiraterone at that time​. Likewise, ADT + enzalutamide or apalutamide has shown a significant survival benefit (about a 33% reduction in risk of death) in clinical trials​
    pubmed.ncbi.nlm.nih.gov. For example, in the ENZAMET trial, adding enzalutamide improved survival (hazard ratio ≈0.67) compared to ADT alone​ pubmed.ncbi.nlm.nih.gov. These combinations also delay disease progression: patients live longer before the cancer spreads or symptoms worsen​ onclive.com. In non-metastatic castration-resistant cases (rising PSA but no visible spread), enzalutamide, apalutamide, or darolutamide with continued ADT greatly delay metastasis by over two years on average and improve overall survival as well​ pubmed.ncbi.nlm.nih.gov.
    Common side effects: AR pathway inhibitors have side effects on top of ADT. Abiraterone can cause joint/muscle pain, high blood pressure, low potassium, swelling, and liver enzyme changes due to excess mineralocorticoids (these are managed by the prednisone given with it)​ aacr.org. The AR receptor blockers like enzalutamide/apalutamide can cause fatigue, weakness, high blood pressure, diarrhea, and sometimes rash or thyroid changes (apalutamide). Enzalutamide (less so darolutamide) can rarely cause seizures or memory trouble, and darolutamide tends to have fewer central nervous system side effects (like less fatigue or dizziness) because it penetrates the brain less. All can increase fall risk in older patients. Despite these side effects, the benefit in survival is significant, and overall quality of life on combination therapy has been shown to be as good as or better than on ADT alone​.

Chemotherapy (Docetaxel and Cabazitaxel)

Mechanism: Chemotherapy works by killing rapidly dividing cells. Docetaxel (Taxotere) is a taxane chemotherapy that interferes with microtubules, causing cancer cells to stop dividing and die. It can attack prostate cancer cells even when they are not as dependent on hormones. Cabazitaxel (Jevtana) is a related chemo drug used if the cancer progresses after docetaxel – it was designed to overcome resistance to first-line chemo.
Administration: Docetaxel is given as an intravenous (IV) infusion, typically every 3 weeks for about 6 cycles (around 18 weeks total). It’s often combined with prednisone pills to reduce inflammation. Cabazitaxel is also IV, usually every 3 weeks, and is used in castration-resistant prostate cancer after other treatments.
Outcomes: Adding docetaxel chemotherapy to ADT in metastatic hormone-sensitive patients was a game-changer. In the CHAARTED trial, men who received ADT + docetaxel lived more than a year longer than those on ADT alone​ajmc.com. Median overall survival was about 57.6 months with the combo vs. 44.0 months with ADT aloneajmc.com. That’s an improvement of 13.6 months (hazard ratio ~0.61)​ ajmc.com. The benefit was especially seen in men with high-volume disease (many bone metastases or organ metastases) – in that subgroup the combo raised median survival to ~51 months vs. 34 months​. This established ADT+docetaxel as a standard for aggressive metastatic cases. For castration-resistant prostate cancer (mCRPC), docetaxel was shown (in earlier trials) to improve survival by a few months compared to older chemo, and it remains an important option if AR-targeted pills stop working. Cabazitaxel, used after docetaxel, can further extend survival. In the CARD trial, cabazitaxel gave a median OS of 13.6 months vs. 11.0 months if patients were switched to another AR therapy instead, a significant improvement​ nvmodagen.nl. Cabazitaxel also improves cancer control (PSA reduction, tumor shrinkage) in those who have already had chemo and ARPIs.
Common side effects: Chemotherapy side effects happen because it also affects normal fast-growing cells. With docetaxel or cabazitaxel, common effects include hair loss, nausea, loss of appetite, fatigue, and neuropathy (numbness or tingling in fingers/toes). It can lower blood cell counts, leading to infection risk (from low white cells) or anemia. Patients often receive medications to prevent nausea and may get growth factor shots if white blood cells drop. Close monitoring by the oncology team is essential​ urmc.rochester.edu. Side effects like fatigue can be helped by staying active (even simple walks) and proper nutrition​ urmc.rochester.edu. Chemo can also cause some memory or concentration issues (“chemo brain”) and nail changes. Cabazitaxel in particular may have slightly more bone marrow suppression (neutropenia); preventive use of growth factors or dose adjustments might be used. Despite these side effects, chemo’s benefits can be important for aggressive disease, and side effects are temporary and manageable with supportive care​.

Targeted Radiotherapy (Radiopharmaceuticals)

These are medicines that deliver radiation directly to cancer cells. They are given by injection and travel in the blood to where the cancer is, releasing radiation to kill tumor cells.

  • Radium-223 (Xofigo): Radium-223 is an injectable radioactive isotope that behaves like calcium, so it naturally homes to bone (where prostate cancer often spreads). It gives off high-energy alpha particles over a short range, killing cancer in the bone but sparing most nearby normal cells.
    Administration: Radium-223 is given as an IV injection, typically once a month for 6 doses. It specifically treats metastatic castration-resistant prostate cancer (mCRPC) that has spread to bones (and is causing symptoms) but not to other organs.
    Outcomes: In the ALSYMPCA phase III trial, radium-223 improved median overall survival to 14.9 months vs. 11.3 months for men with mCRPC that had spread to bones​ pubmed.ncbi.nlm.nih.gov. That’s roughly a 30% reduction in the risk of death. It also delayed complications like fractures or spinal cord compression. Uniquely, radium-223 can reduce bone pain significantly, improving quality of life. It’s now an approved standard option for bone-predominant CRPC.
    Common side effects: The main side effects are bone marrow suppression (low blood counts, especially low platelets or white cells) and mild GI upset (nausea, diarrhea). Unlike external radiation, radium-223 usually does not cause hair loss or severe sickness. Some patients may feel increased bone pain briefly (flare) when treatment starts, but this is usually temporary. Regular blood tests are required to monitor counts. Radium-223 is generally well tolerated.

     

  • Lutetium-177 PSMA (Pluvicto): Lutetium-177 PSMA is a newer targeted radiotherapy that was approved in 2022. It is a radioactive molecule that seeks out PSMA (a protein on prostate cancer cells) and delivers beta radiation. A small targeting molecule (PSMA ligand) carries the radioactive Lutetium-177 directly to prostate cancer cells anywhere in the body that express PSMA.
    Administration: Given by IV infusion every 6 weeks (for up to 6 cycles), after confirming the cancer has PSMA uptake on a specialized PET scan. It’s used for mCRPC patients who have already been treated with ARPIs and chemotherapy.
    Outcomes: In the VISION trial, Lu-177 PSMA therapy plus standard care improved survival to a median of 15.3 months vs. 11.3 months with standard care alonepmc.ncbi.nlm.nih.gov. This ~4-month benefit was significant in men who had exhausted other treatments. Many patients also showed tumor and PSA reductions. Ongoing studies are testing Lu-177 PSMA in earlier disease and in combination with other treatments (potentially even better results).
    Common side effects: Dry mouth (salivary glands can take up some PSMA tracers), nausea, fatigue, and bone marrow suppression can occur. The radiation can temporarily lower blood counts (anemia, low white cells or platelets). Patients are monitored for these and for kidney function (as it is cleared through kidneys). Overall, it’s fairly well tolerated for most; side effects are generally mild to moderate.

     

Immunotherapy (Stimulating the Immune System)

Prostate cancer has been challenging for traditional immunotherapy, but one FDA-approved immunotherapy is sipuleucel-T (Provenge). Others, like checkpoint inhibitors, are used in select cases.

  • Sipuleucel-T (Provenge): This was the first cancer vaccine approved. It is a personalized immunotherapy made from a patient’s own immune cells. The process involves drawing blood to collect immune cells (dendritic cells), exposing them to a prostate cancer protein (PAP) plus an immune stimulant in the lab, and then infusing them back into the patient. The goal is to “train” the immune system to attack prostate cancer cells.
    Administration: It is given in three IV infusions, spaced two weeks apart (each dose is custom-made from the patient’s cells). It’s typically used for asymptomatic or minimally symptomatic mCRPC (castration-resistant, metastatic disease) before or in lieu of chemotherapy.
    Outcomes: In the pivotal IMPACT trial, sipuleucel-T improved 3-year survival rates and gave a median overall survival about 4.1 months longer than placebo (25.8 vs 21.7 months)​ aacrjournals.org. While it usually does not cause PSA drops or tumor shrinkage on scans, it can prolong life. The idea is that it works in an “invisible” way by priming T-cells to slow the cancer’s growth. Real-world registry data have shown median survival over 30 months for patients receiving sip-T in routine practice​ pmc.ncbi.nlm.nih.gov. Sipuleucel-T is an approved standard option for mCRPC with low symptom burden.
    Common side effects: Because it’s an immune infusion, the main side effects happen within a day of infusion: fever, chills, fatigue, nausea, and headache – like flu-like symptoms. These usually last only a day or two and are managed with acetaminophen or ibuprofen. There is a small risk of infusion reactions (rash, breathing issues) but severe reactions are rare. Overall, side effects are mild, and recovery after each infusion is quick, making it generally well tolerated.

     

  • Checkpoint Inhibitors (Pembrolizumab): Standard immunotherapy drugs (like PD-1 inhibitors) have had limited success in prostate cancer, except in certain cases. Pembrolizumab (Keytruda) is FDA-approved not specifically for prostate cancer but for any solid tumor with high microsatellite instability (MSI-H) or DNA mismatch repair deficiency – about 2–3% of prostate cancers have this feature. In those rare cases, pembrolizumab can cause dramatic tumor responses. Outside of that genetic subset, single-agent immunotherapy helps only a small proportion of prostate cancer patients​. Researchers are actively studying combinations (e.g., pembrolizumab plus enzalutamide or plus chemotherapy) to see if immunotherapy can be made more effective. Common side effects: Checkpoint inhibitors can cause immune-related side effects – inflammation in normal organs (like skin rash, colitis, liver issues, lung inflammation). These drugs are used only for select patients, so we won’t focus too much here as they are not standard for most advanced prostate cancers.

     

PARP Inhibitors and Other Targeted Therapies

Some advanced prostate cancers have specific genetic mutations that can be targeted by newer drugs:

  • PARP Inhibitors (Olaparib, Rucaparib): These are pills that block PARP, an enzyme that helps repair DNA damage in cells. They are particularly effective in cancers that already have DNA-repair defects (like mutations in BRCA1 or BRCA2 genes). When a prostate cancer has a BRCA mutation, blocking PARP causes so much DNA damage that the cancer cells die.
    Administration: Olaparib (Lynparza) and Rucaparib (Rubraca) are taken as oral tablets or capsules, usually twice daily. They are approved for mCRPC patients who have specific DNA-repair gene mutations (such as BRCA1/2, ATM, etc.) and whose cancer has progressed after AR-targeted therapy and (for rucaparib) chemotherapy.
    Outcomes: In men with BRCA-mutated mCRPC, PARP inhibitors can significantly shrink tumors and prolong survival. For example, the PROfound trial showed olaparib improved survival and delayed progression versus standard AR therapy in these patients​ aacr.org. Recently (2023), the FDA approved olaparib + abiraterone in combination as an initial therapy for mCRPC with BRCA1/2 mutations​. In a trial, among patients with BRCA mutations, those who got olaparib + abiraterone had a much longer time before progression (median not reached, vs 8 months on abiraterone alone) and were 70% less likely to die during the study than those on abiraterone alone​. This shows a major benefit in that subgroup. (Notably, in patients without BRCA mutations, adding olaparib did not give a significant survival benefit​, so this combination is targeted to the right patients.) Rucaparib has also shown high response rates in BRCA-mutated prostate cancer and is approved after other treatments.
    Common side effects: PARP inhibitors can cause anemia and fatigue (due to effects on bone marrow), as well as nausea, decreased appetite, and GI upset. Some patients get altered taste or cough. These side effects are generally manageable with dose adjustments. Regular blood counts are done to check for anemia.

     

  • Other targeted therapies: A small portion of prostate cancers have mutations in genes like PTEN/AKT or others. Clinical trials in the last few years have explored drugs for these. For instance, an AKT inhibitor called capivasertib showed promise in a Phase III trial (CAPItello-281) for metastatic prostate cancer with PTEN loss. When added to abiraterone and ADT in newly diagnosed metastatic cases, capivasertib significantly improved radiographic progression-free survival (how long patients live without the cancer growing)​. There was also an early trend toward better overall survival with this combination​. As of 2025, capivasertib is not yet standard, but it may become an approved option for PTEN-deficient cancers in the near future.

     

All these approved treatments can be combined in various ways depending on the situation. For instance, a common approach for metastatic hormone-sensitive prostate cancer is ADT + one of the AR pathway inhibitors, or ADT + chemotherapy, or even ADT + AR inhibitor + chemotherapy for very aggressive cases. For castration-resistantdisease, doctors may sequence multiple treatments: AR inhibitors, then chemo, then targeted radiotherapy, etc., or even use two together (like the newly approved olaparib+abiraterone for those with BRCA mutations). The goal is to hit the cancer from different angles – lower hormones, block hormone use, kill cells directly, target bone mets, and so on – to improve survival.

2. Comparison Table: Treatment Effectiveness vs. Side Effects

The following table summarizes major treatment options for advanced prostate cancer, focusing on how effective they are (especially at extending life) and their notable side effects. This comparison assumes these treatments are added on top of standard ADT (since ADT is the backbone for advanced disease). “Effectiveness” here is described in terms of overall survival (OS) improvement or risk reduction in clinical studies, and “Side Effects” highlights the most common or significant issues to watch for.

Treatment (Combination) Effectiveness (Overall Survival Benefit) Common Side Effects
ADT alone(androgen deprivation therapy) Baseline therapy – controls cancer temporarily, but cancer usually progresses in ~1–2 years on ADT alone (no added survival beyond historical norms). 5-year survival with hormone-sensitive mets <50%, and for castration-resistant mets only ~30%​. Hot flashes, reduced libido/ED, fatigue, weight gain, muscle loss, bone thinning (osteoporosis), mood changes.
ADT + AR Pathway Inhibitor (e.g. Abiraterone, Enzalutamide, Apalutamide, Darolutamide) Significantly improves survival: Reduces risk of death by ~30–40% vs ADT alone​ pubmed.ncbi.nlm.nih.govAbiraterone + ADT: 3-year survival ~66% vs 49% on ADT​ (median OS ~ not reached vs 34.7 mo)​.
Enzalutamide/Apalutamide + ADT: Similar ~33% lower death risk​ pubmed.ncbi.nlm.nih.gov; markedly prolongs time to progression (often adds years). Proven benefit in both metastatic and non-metastatic CRPC settings (enzalutamide/apalutamide/darolutamide delay metastasis by ~2 years and improve OS).
Fatigue, hypertension (high blood pressure), mild nausea, diarrhea.
Abiraterone: can cause fluid retention, high blood pressure, low potassium (due to increased mineralocorticoids)​; requires prednisone.

Enzalutamide/Apalutamide: Fatigue, weakness, risk of falls; enzalutamide can cause rare seizures.
Darolutamide: similar to enzalutamide but typically less fatigue and CNS effects.

ADT + Docetaxel(chemotherapy) Improves survival by ~13–15 months in fit patients with metastatic hormone-sensitive disease ajmc.com. Median OS ~57.6 mo with ADT+docetaxel vs 44.0 mo ADT alone​

(HR ~0.61). Greatest benefit in high-volume metastases (e.g. 51 mo vs 34 mo)​. In castration-resistant disease, docetaxel also improves survival by a few months vs no chemo (established in earlier trials).

Hair loss, nausea, fatigue, low blood counts(infection risk), neuropathy (numb/tingling in hands/feet), nail changes. Needs monitoring of blood counts. Side effects are temporary and manageable with supportive meds urmc.rochester.edu (e.g. anti-nausea drugs, growth factors).
ADT + AR Inhibitor + Docetaxel(“Triplet” therapy for high-risk disease) Largest survival benefit seen for very aggressive cases. The addition of darolutamide (an AR inhibitor) to ADT+docetaxel improved OS by ~32% (HR = 0.68) vs ADT+docetaxel alone​. At 4 years, more patients were alive with the triplet (e.g. 62% vs 50% in one analysis). Triplet therapy is now recommended for metastatic hormone-sensitive patients with high-volume or high-risk features. Combined side effects of hormone therapy + chemo. Expect those listed for ADT (hormonal side effects) plus those from chemotherapy. Notably: fatigue can be more pronounced when AR inhibitors and chemo are given together. Needs close monitoring, but studies show quality of life is not significantly worse with the combo​ if side effects are well-managed.
Radium-223(alpha radiotherapy for bone mets) Extends survival in metastatic CRPC with bone metastases (median OS ~14.9 vs 11.3 mo placebo) pubmed.ncbi.nlm.nih.gov. Provides effective pain relief in bone metastases and delays skeletal complications (fractures). Best for patients with multiple painful bony metastases and no visceral mets. Bone marrow suppression (can cause low blood counts, especially platelets). Mild nausea, diarrhea. Usually well tolerated; low risk of hair loss or other classic chemo effects. Monitor blood counts regularly.
Lutetium-177 PSMA (targeted radioligand) Improves survival in advanced mCRPC after other treatments (median OS ~15.3 vs 11.3 mo on standard care)​ pmc.ncbi.nlm.nih.gov. Often dramatically lowers PSA and shrinks tumors in PSMA-positive patients. Clinical trials show improved progression-free survival and quality of life (delayed pain progression) compared to non-targeted care. Dry mouth (from salivary gland radiation), fatigue, nausea. Can cause anemia or low platelets after several doses (radiation to bone marrow). Generally moderate side effects; some patients may need blood transfusions if counts drop.
Sipuleucel-T(immunotherapy vaccine) Modestly improves survival (adds ~4 months median life extension​ aacrjournals.org; 3-year survival 32% vs 21% in trial). Does not shrink tumors or lower PSA much, but prolongs lifespan in asymptomatic mCRPC. Best used early in mCRPC before heavy symptoms or chemo. Infusion reactions: fever, chills, flu-like symptomswithin a day of treatment (usually mild and brief). Headache, fatigue, nausea possible after infusions. Very low risk of serious events; generally well tolerated.
PARP Inhibitor + ABi (e.g. Olaparib + Abiraterone) (for BRCA-mutated mCRPC) Significant benefit in BRCA1/2-mutated prostate cancer: In BRCA-mutant patients, combo showed much longer time to progression (median not reached vs 8 months) and a 70% reduction in risk of death vs abiraterone alone. Leading to FDA approval in 2023 for BRCA+ mCRPC​. (No proven OS benefit in non-BRCA patients​ aacr.org.) Anemia and fatigue (due to olaparib). Nausea, loss of appetite, and GI issues (olaparib) on top of abiraterone’s side effects (high blood pressure, low potassium, etc.). Need periodic blood counts (for anemia). Most side effects are manageable with dose adjustments.
Checkpoint Inhibitor (Pembrolizumab)(for MSI-high tumors only) Effective only in a small subset of prostate cancers. In MSI-high/mismatch repair deficient tumors (~3% of patients), can induce long-lasting tumor regression in about half of those cases. Not beneficial in routine prostate cancer (trials show low response in unselected patients​). Immune-mediated side effects: can cause inflammation of healthy organs – e.g. skin rash, colitis (diarrhea), liver inflammation, lung issues – which may require steroid treatment. Used only in select patients due to these risks and low general efficacy in prostate cancer.

3. Prognosis and Survival Rates in Advanced Prostate Cancer

How long can someone live with advanced prostate cancer? The answer depends on many factors: the extent of disease (stage), the treatments used, tumor biology (aggressiveness), patient health and comorbidities, and even race/ethnicity and access to care. Here we summarize survival statistics and what influences prognosis, based on recent research (mostly from the last 7 years):

  • By Disease Stage: Survival is much better when prostate cancer is diagnosed earlier. For localized prostate cancer, 5-year survival is nearly 100%. However, once the cancer is metastatic (spread to distant sites), the historical 5-year relative survival drops to around 30%​. This means only about 1 in 3 men with metastatic prostate cancer traditionally survive 5 years. That said, new combination therapies have improved these odds in recent years. For example, men with metastatic hormone-sensitive prostate cancer now often live well beyond 3–5 years with the use of early intensive treatment (ADT plus other agents). Clinical trial populations show median survivals of 4–5 years when therapies like abiraterone or docetaxel are started up front​. Some patients, especially with low-volume metastatic disease and effective treatment, can live much longer (5–10 years). In castration-resistant metastatic prostate cancer (mCRPC) – which is more advanced – median survival used to be ~18 months in the early 2000s; with the multiple newer treatments available, many patients now live 2–3+ years even after developing resistance, especially if they can receive several lines of effective therapy sequentially. It’s important to note these are medians – individual outcomes vary widely.

     

  • Impact of Treatment Type: Using combination treatments early can significantly improve survival. Multiple trials have shown that men who receive treatments like ADT + AR inhibitors or chemo when the disease is still hormone-sensitive live longer than those who wait to use those therapies until later​. For instance, starting abiraterone or enzalutamide at the metastatic hormone-sensitive stage can yield 5-year survival rates on the order of 60–70% in some trial subsets, versus perhaps 30–40% if just ADT was used and these drugs were delayed. Similarly, the recent triplet therapy (ADT+docetaxel+darolutamide) led to an estimated 4-year survival rate of about 62%, compared to 50% with ADT+docetaxel alone​ urotoday.com. In mCRPC, adding drugs like olaparib for BRCA-mutated cancer or Lu-177 PSMA after other treatments can extend life by months to a year or more on average pmc.ncbi.nlm.nih.gov. Therefore, prognosis is improving as new combinations are used: it’s not unheard of now for some patients with metastatic disease to live 5-8 years or longer with a sequence of modern treatments, especially if they respond well to each line of therapy.

     

  • Comorbidities and Age: A patient’s overall health affects prognosis. Someone with serious other conditions (like heart disease, uncontrolled diabetes, etc.) may not tolerate aggressive treatments or may have their life expectancy limited by those illnesses. Conversely, a healthy patient in his 50s or 60s with metastatic prostate cancer might do very well with aggressive treatment. Clinical trials often report better outcomes in patients who are fit enough to receive all therapies. For example, older frail patients might not receive chemotherapy due to risks, which could influence their survival. However, even men in their 80s can often take AR pathway inhibitors and benefit. Doctors tailor therapy intensity to a patient’s health – a balance between extending life and maintaining its quality.

  • Racial and Ethnic Disparities: Prostate cancer outcomes differ by race, due to a mix of biology and access to care. Historically, African American men have had higher prostate cancer death rates than White men (more than twice as likely to die of the disease)​health.harvard.edu. They often present with more advanced disease, possibly from disparities in healthcare access and other factors. However, when treated with equivalent therapy, outcomes can be similar or even better in African American men compared to others​ health.harvard.edu. Recent research pooling clinical trial data found that African American and White patients had similar survival on treatments like docetaxel (about 21 months median in mCRPC), and after adjusting for other factors, African American men were 20% less likely to die during those trials​. Another VA study (equal-access setting) showed African American men with metastatic cancer lived 30 months on novel hormonal therapy vs 26 months for White men​. Scientists are investigating whether certain genetic factors or immune responses might give African American patients better response to some treatments​. The key takeaway is that equitable access to standard-of-care treatments can greatly reduce survival disparities​. All patients, regardless of race, should be offered the effective combinations discussed – when that happens, survival gaps close and can even reverse in some studies. Unfortunately, outside clinical trials, disparities persist: African American men and other minorities may have less access to specialized care or new therapies, affecting outcomes. Efforts are ongoing to ensure everyone gets timely, appropriate treatment to improve prognosis.

     

In summary, survival rates for advanced prostate cancer are improving due to combination treatments. Many men with newly diagnosed metastatic prostate cancer can expect to live several years, especially if they receive modern therapies early. For instance, a man with metastatic hormone-sensitive prostate cancer treated with ADT plus abiraterone might have a median survival of well over 4–5 years​, whereas a decade ago on ADT alone it might have been 3 years or less. If the cancer becomes castration-resistant, median survival may still be 2–3 years or more with successive treatments (chemo, other AR inhibitors, PARP therapy, radioligand, etc.). There are of course cases that do better (long-term responders) and cases that do worse (aggressive biology that resists treatment). Doctors often refrain from giving an exact “time left” prognosis because it varies so much. Instead, they focus on treating aggressively while maintaining quality of life, and new research from the last 7 years gives hope that outcomes will continue to get better.

 

4. Emerging Therapies on the Horizon

Research in advanced prostate cancer is very active. Many new treatments and combinations are being tested in clinical trials (Phases I, II, and III). Here are some of the most promising emerging therapies (especially those in Phase III or showing strong Phase I/II results) as of 2025:

  • Next-Generation Hormonal Agents: While we already have potent AR pathway inhibitors, researchers are developing even more advanced ways to shut down androgen signaling. One approach is PROTACs and AR degraders, which are drugs that cause the destruction of the androgen receptor protein. Several AR degraders (like ARV-110) are in early trials. These aim to work even if cancer becomes resistant to enzalutamide or apalutamide. Early results have shown some PSA reductions in heavily pretreated patients, but research is ongoing.

  • AKT Pathway Inhibitors: As mentioned, cancers with loss of the PTEN gene (common in ~40-50% of advanced cases) activate the AKT pathway to grow. In addition to capivasertib (which in a Phase III trial significantly improved progression-free survival when added to standard therapy for PTEN-deficient cancers​), another drug ipatasertib was tested in a trial (IPATential) with mixed results (it helped in the subset with PTEN loss, but not overall). The success of capivasertib means we may soon have a targeted drug for this subgroup. These drugs would be combined with AR therapy to maximize benefit.

     

  • Combination Immunotherapy: Pure immunotherapy alone (like single checkpoint inhibitors) hasn’t been a breakthrough for most prostate cancer patients. But combining immunotherapy drugs or pairing them with other treatments could change that. Checkpoint inhibitor combinations are being tested – for example, nivolumab (PD-1 inhibitor) + ipilimumab (CTLA-4 inhibitor) in mCRPC (CheckMate 650 trial) showed some tumor responses, though with high side effect rates. Researchers are refining doses to improve safety. Another strategy is combining pembrolizumab with enzalutamide (KEYNOTE-199 cohort) or pembrolizumab with chemotherapy(KEYNOTE-921) – early data suggest only modest improvements, but certain patients (like those with PD-L1 expression or other immune markers) might benefit​ pubmed.ncbi.nlm.nih.gov. Trials are also looking at immunotherapy after radiation or Lu-177 (since radiation can make tumors more visible to the immune system). While no new checkpoint immunotherapy combo is standard yet, these trials are important and ongoing.

     

  • Bispecific T-Cell Engagers (BiTEs) and CAR-T Cells: These are highly innovative immune therapies. BiTEs are artificial antibodies that bind a T-cell on one side and a cancer cell (via a target like PSMA) on the other side, bringing them together so the T-cell can kill the cancer. An example is AMG 160, a bispecific antibody targeting PSMA on prostate cancer and CD3 on T-cells. In a Phase I trial, AMG 160 showed PSA declines in ~69% of patients (34% had >50% PSA reduction) in very advanced mCRPC​. However, many patients had cytokine release syndrome (fever, immune reaction) that needs to be managed​. This is very promising because it suggests the immune system can be harnessed if we can control the side effects. CAR-T cell therapy (genetically engineering a patient’s T-cells to attack a prostate cancer antigen) is also under study. Some early CAR-T trials against PSMA or STEAP1 (another prostate antigen) have shown sporadic successes but also risks (like severe inflammatory reactions). These immune therapies are mostly in Phase I/II, but they represent a potential leap forward if they become safer and more effective.

     

  • Novel Radiopharmaceuticals: Building on the success of Radium-223 and Lu-177 PSMA, researchers are exploring other radioactive compounds. One exciting area is actinium-225 (Ac-225) labeled to PSMA-targeting ligands. Actinium-225 emits alpha particles (like a super potent, short-range blast) which may kill cancer cells more effectively than beta particles. Early reports (Phase I) of Ac-225 PSMA therapy have shown very high PSA response rates, even in cancers that resisted Lu-177, but also some significant side effects like dry mouth and marrow suppression. Efforts are ongoing to optimize dosing. Also, combinations of radioligand therapy with other treatments are being tried: for example, trials are testing Lu-177 PSMA + immunotherapy or Lu-177 PSMA + PARP inhibitor to see if they work better together (the rationale is radiation may make cancer cells more susceptible to PARP inhibitors or more visible to immune cells).

  • New Targeted Drugs: Aside from PARP and AKT, scientists are targeting other vulnerabilities. One target is HSP-27 (heat shock protein) which helps cancer cells survive treatment stress – an HSP27 inhibitor (apatorsen) was tested but didn’t significantly prolong survival in mCRPC. Another target is c-Myc or other oncogenes – still mostly preclinical. Angiogenesis inhibitors (like bevacizumab, which cuts off blood supply) largely failed in prostate cancer trials, but combinations with them are being revisited with modern immunotherapy to see if there’s synergy. There are also vaccines like PROSTVAC (a PSA-targeted vaccine) that in Phase II looked promising but a Phase III trial did not improve survival; however, it’s now being tested in combinations (e.g., with checkpoint inhibitors) where it might work better.

  • Epigenetic and Precision Medicine Approaches: Advanced prostate cancers sometimes undergo neuroendocrine differentiation (an aggressive form) after many treatments. Researchers are looking at drugs to target neuroendocrine prostate cancer (which behaves more like small-cell cancer). Additionally, more comprehensive genomic testing of tumors is becoming common, and this could identify rare targets – for example, some prostate cancers have NTRK fusions (targetable by TRK inhibitors), or high tumor mutational burden (which might respond to immunotherapy). These situations are rare but exemplify precision medicine – matching a specific drug to a specific mutation.

In summary, the pipeline for advanced prostate cancer includes more personalized treatments (based on genetic mutations like BRCA or PTEN status), more potent hormonal therapies, immune-based treatments that might finally overcome prostate cancer’s resistance to immunotherapy, and better targeted radiation. Some of these emerging therapies (like PARP inhibitor combinations and PSMA radioligands) have already moved into practice, while others (like BiTEs, CAR-T cells, and next-gen targeted drugs) are likely a couple of years away but could dramatically change the landscape. It’s an evolving and hopeful field – every year, new data comes out at oncology conferences (ASCO, ESMO, etc.) that refine how we treat advanced prostate cancer. Patients are encouraged to ask about clinical trials as well, because accessing these emerging therapies through trials can sometimes offer cutting-edge options if standard treatments are no longer effective.

5. Managing Side Effects and Recovery

Treatments for advanced prostate cancer, while life-prolonging, can bring a range of side effects. Managing these side effects is crucial for maintaining quality of life. Below are practical strategies to handle common side effects, and tips for recovery during and after treatment:

  • Fatigue: Fatigue is one of the most common side effects, whether from hormone therapy, chemotherapy, or radiation​. It’s a feeling of extreme tiredness that doesn’t go away with rest. To combat fatigue, studies show that staying physically active helps significantly. Even gentle exercise like daily walking, yoga, or light resistance training can boost energy and reduce fatigue​. Set small activity goals – for example, a short walk in the morning when energy tends to be highest. Good sleep hygiene is important: maintain a regular sleep schedule and ensure a restful environment. Eating a balanced diet and staying hydrated also help with energy levels. If anemia (low red blood cells) is causing fatigue (common with chemo or PARP inhibitors), treating the anemia (with iron, erythropoietin, or transfusions if needed) can improve symptoms. Some patients find integrative therapies like acupuncture, massage, or meditation useful for fatigue as well​. It’s key to communicate severe fatigue to your doctor – occasionally, medication doses can be adjusted or physical therapy consulted to help.

     

  • Hot Flashes and Sweats: These are typically from ADT (lowering testosterone). They can range from mild warmth to drenching sweats and are similar to menopausal hot flashes. Lifestyle measures: dress in layers, keep cool at night with a fan, avoid triggers like spicy food or hot drinks. If very bothersome, doctors can prescribe medications: low-dose megesterol acetate, certain antidepressants (SSRIs or SNRIs), or gabapentin have all been shown to reduce hot flash frequency. Some men get relief from acupuncture as well. Over time, the body does adjust and hot flashes may become less intense.

  • Sexual Dysfunction: ADT causes decreased libido and erectile dysfunction (ED) in most men. This can be emotionally challenging. Open communication with your partner and doctor is important. For ED, treatments like PDE5 inhibitors (Viagra/sildenafil, Cialis/tadalafil), vacuum erection devices, or penile injections can be considered – though their effectiveness may be limited during ADT, they can still help some. If sexual activity is a priority, discussing intermittent ADT (if appropriate) or early use of erectile aids can be useful. Counseling or sex therapy might help couples adjust to changes in intimacy. Remember that it’s a common side effect and there are support groups for men dealing with these issues.

  • Bone Health and Pain: Long-term ADT can weaken bones (osteoporosis), and metastases to bone can cause pain or fractures. To maintain bone health, ensure adequate calcium and Vitamin D intake (through diet or supplements). Weight-bearing exercise (like walking or light weightlifting) strengthens bones and muscles. Doctors often prescribe bone-protecting agents such as bisphosphonates (zoledronic acid) or denosumab (Xgeva) for men with bone metastases or long-term ADT use – these medications strengthen bones and reduce fracture risk. For bone pain from metastases, radiation therapy to painful spots or the use of radiopharmaceuticals (like radium-223) can provide significant relief. Pain medications, from NSAIDs to opioids, should be used as needed – there is no benefit to “toughing out” cancer pain; controlling pain can improve function and even survival. If you’re on long-term steroids (like prednisone with abiraterone), make sure bone health is monitored closely as steroids also thin bones.

  • Nausea and Appetite Loss: Chemotherapy or radioligand therapy can cause nausea. Proactively using anti-nausea medications (ondansetron, dexamethasone, etc.) before and after treatments is very effective​. If one antiemetic doesn’t work well, there are several others to try – so let your team know if you still feel nauseous. Eating small, frequent bland meals, and staying hydrated with fluids like ginger tea or electrolyte drinks can help. Some patients find relief with ginger supplements or acupressure bands. Appetite loss can be countered by focusing on nutrient-dense foods in whatever quantity you can handle. Sometimes appetite stimulants (like megestrol) are prescribed, but these have to be weighed against risks (megestrol can increase clot risk). Involving a nutritionist can provide personalized suggestions.

     

  • Diarrhea or Digestive Issues: Certain AR inhibitors (apalutamide, enzalutamide) and chemo can cause loose stools. Anti-diarrheal medications like loperamide (Imodium) can be used as needed. Follow the BRAT diet (Bananas, Rice, Applesauce, Toast) during episodes of diarrhea. Stay hydrated. If a particular medication consistently causes diarrhea, ask about dose adjustments or supportive meds (for example, some patients take fiber supplements to bulk up stool). Conversely, pain meds can cause constipation, so if you’re on opioids for pain, use stool softeners or mild laxatives preventively.

  • Hand-Foot Syndrome or Neuropathy: Docetaxel can cause neuropathy (numbness/tingling in extremities). While there’s no guaranteed prevention, keeping hands and feet warm (but not hot) and avoiding pressure can help. Report neuropathy early – if it worsens, dose adjustments might be needed to prevent permanent nerve damage. Some patients find supplements like B vitamins or glutamine helpful, though evidence is limited. After chemo, neuropathy usually improves slowly over months. Maintaining gentle exercise of hands/feet (like squeezing a stress ball, walking) can maintain circulation and function.

  • Mood Changes and Cognitive Effects: ADT can cause mood swings, irritability, or even depression. Additionally, the stress of dealing with advanced cancer can understandably affect mental health. It’s important to address this: consider joining a support group (many prostate cancer organizations and hospitals host support groups for patients and caregivers). Talking to a counselor or psychologist, especially one familiar with cancer, can provide coping strategies. If depression or anxiety is significant, medications like antidepressants or anti-anxiety meds may be useful – there is no shame in needing these, as this is a challenging journey. “Chemo brain” (mild cognitive impairment during/after chemo) is usually temporary; mental exercises, keeping a planner, and giving yourself grace with memory lapses help. Involve loved ones in helping manage complex tasks during intensive treatment periods.

  • Recovery and Rehabilitation: After treatments like chemotherapy or a series of radiation infusions, your body may need time to recover. Schedule rest periods and don’t overextend yourself. Physical therapy or exercise programs tailored for cancer survivors (like cancer rehab or “prehab” programs) can be very beneficial – they help you regain strength, improve endurance, and reduce treatment-related fatigue or deconditioning. Many cancer centers offer such programs. Pelvic floor therapy can help if you experience urinary incontinence (for instance, if you had prostate surgery or radiation affecting control). If lymphedema (leg swelling) occurs after lymph node treatments, there are lymphedema therapists who can assist. Essentially, utilize rehabilitation services to address any specific functional issues.

  • Routine Monitoring: During any treatment, you will have regular check-ups and lab tests. Keep a symptom diary to track side effects and discuss them at visits. Your healthcare team can then adjust doses or add supportive treatments accordingly. For example, if ADT gives you significant fatigue and your testosterone is adequately suppressed, sometimes they might space ADT shots a bit further apart under close monitoring. Or if an ARI is causing high blood pressure, adding an antihypertensive medication can control that.

  • Lifestyle Factors: Don’t underestimate general healthy habits. Nutrition: Aim for a balanced diet rich in fruits, vegetables, lean protein, and whole grains. Omega-3 fatty acids (like from fish or flaxseed) might help with inflammation and heart health. Limit excessive sugar and processed foods. Avoid tobacco and moderate alcohol – smoking can worsen outcomes and interfere with healing, and alcohol can exacerbate certain side effects (and should be limited especially if on certain meds or with liver metastases). Hydration: Drink plenty of fluids, especially if on treatment causing diarrhea or if you’re not eating a lot; this prevents dehydration and helps kidneys flush out drug byproducts.

  • Family and Social Support: Side effects can be tough to handle alone. Accept help from family or friends – whether it’s rides to appointments, help with meals, or just someone to talk to. Social support has been shown to improve coping and even outcomes for cancer patients. If you feel you’re a burden (a common worry), remember that loved ones generally want to help, and letting them can make them feel involved in your care.

  • Communication with Healthcare Team: Always inform your oncologist or nurse about side effects you experience. They expect these side effects and have many ways to help. For example, if you have severe neuropathy, they might pause chemotherapy to let nerves recover, rather than continuing until it’s debilitating. If you have mood issues, they can refer you to psycho-oncology services. There are also palliative care specialists – not just for end-of-life, but during treatment – who focus on symptom management (pain, nausea, etc.). Early palliative care involvement in metastatic cancer has been shown to improve quality of life and even survival in some studies, by aggressively managing symptoms. Utilizing these resources is a sign of good, holistic care.

In essence, managing side effects is about being proactive and communicative. With modern supportive medications and integrative therapies, most treatment side effects can be lessened to a degree that patients can continue daily activities. Recovery is an ongoing process – even as treatment continues, your body can adapt and recuperate with the right support. After finishing a treatment course (like a round of chemo), give yourself time to bounce back, and celebrate the milestone. Many men live for years on continuous or intermittent therapy for advanced prostate cancer, so prioritizing your overall wellness alongside cancer control leads to the best outcomes in the long run.

6. Talking to Doctors About Treatment Options

Navigating advanced prostate cancer can be overwhelming. It’s crucial to have open, honest conversations with your healthcare providers. Here are some tips and guidance on how to approach discussions with your doctors (oncologist, urologist, etc.) and ensure you make informed decisions together:

  • Be Open and Honest: From the start, practice clear communication. Feel free to ask any question, no matter how small – doctors are used to it​. If you don’t understand something, ask for clarification. Let your doctor know about all symptoms you’re experiencing (even if you’re not sure they’re related to the cancer or treatment). For example, if you feel depressed or extremely tired, tell them – they can only help with issues they know about.

     

  • Understand Your Specific Disease: Ask your doctor to explain your cancer stage and status. Is it still hormone-sensitive or has it become castration-resistant? How extensive are the metastases? Understanding this helps frame the treatment choices. For instance, “Is my cancer still hormone-sensitive, and what does that mean for my treatment options?”​. If something is unclear (like a medical term in a scan report), ask them to break it down in simpler language.

  • Ask About Treatment Goals: Different treatments may have different goals (some aim to prolong survival, others to reduce symptoms or improve comfort). It’s important to know “What is the goal of this recommended treatment?” Is it to cure (in advanced prostate cancer, treatments are usually not curative but control-oriented), to extend life, to delay progression, or to relieve symptoms? Understanding this helps set expectations. For advanced prostate cancer, the goal is often to extend life while maintaining quality of life as much as possible.

  • Discuss All Options: When deciding on a plan, ask about all your options. For example: “What are the treatment options in my case, and why are you recommending this one first?” There might be choices between chemo now vs later, or between different hormonal therapies. Have the doctor explain the pros and cons of each. You can ask about clinical trials as well: “Are there any clinical trials that I should consider at this stage?” Trials can offer access to emerging therapies and are worth inquiring about, especially if standard options are limited or if you’re interested in contributing to research.

  • Bring Up Quality of Life: Let your doctor know what matters to you personally. Some patients prioritize living as long as possible, others prioritize quality of life or independence. Communicate your values – e.g., “I really want to avoid severe fatigue because I take care of my grandson” or “I’m willing to go through intensive treatment if it gives me a chance to live longer.” This helps your doctor tailor recommendations. It’s a partnership: your goals should guide the treatment strategy alongside the medical indications.

  • Ask About Side Effects and Management: For each proposed treatment, ask: “What are the common side effects, and how can we manage them?” This not only prepares you for what to expect, but also gives you confidence that side effects can be handled. Doctors can tell you which side effects are temporary, which might require meds to control, etc. For instance, if chemotherapy is an option, you might ask, “How will we control nausea and infection risk during chemo?” and the doctor can explain plans for anti-nausea drugs and monitoring blood counts. Knowing there’s a plan for side effects can make the treatment feel less intimidating.

  • Involve Your Support System: Consider bringing a family member or friend to appointments​. They can help listen, take notes, and ask questions you might not think of. Often, two sets of ears are better than one – especially when a lot of information is being discussed. Your companion might remember details that you miss. It’s also helpful emotionally to have someone with you. Doctors are accustomed to patients coming with caregivers, so you won’t be bothering them – it’s usually welcomed.

     
  • Take Notes or Record: Write down key points during the meeting, or ask if you can record the conversation on your phone (so you can replay it later – but always ask permission first). This way, you don’t have to rely on memory alone. Doctors often give a lot of details (names of drugs, scheduling, etc.), and it’s easy to forget. A written summary helps you review and formulate follow-up questions.

  • Ask for Clarification on Prognosis: It’s reasonable to ask, “What is my outlook with these treatments?” Keep in mind prognosis is an estimate. Some patients want detailed statistics; others prefer general answers. Let your doctor know how much you want to know. If you ask, “What’s the typical survival time for someone in my situation?”, be prepared that you might get ranges or probabilities rather than a definite answer. Many doctors answer in terms of “We hope to control this for several years” or “Median survival is about X, but many people do better or worse.”Use this information as a guide, not a guarantee. It can help you plan for the future while maintaining hope.

  • Encourage Shared Decision-Making: If you’re unsure about a treatment, voice it. For instance, “I’m worried about the side effects of chemotherapy – are there alternatives, or what happens if we delay it?” Your doctor can then discuss consequences or alternatives. The trend in cancer care is shared decision-making, meaning your preferences are considered along with the doctor’s expertise. Sometimes there’s a “right” answer medically, but often there are options with different trade-offs, and your input is crucial. Don’t hesitate to speak up if you feel uncomfortable with a plan; there may be flexibility or at least you deserve a clear explanation of why a certain approach is recommended.

  • Ask Specific Questions: It might help to have a list of specific questions. Examples:

    • “What are the benefits and risks of this treatment?”​
    • “How will we know if the treatment is working? What monitoring will we do (PSA tests, scans) and how often?”
    • “If this treatment doesn’t work, what is the next plan?” (Knowing there’s a plan B and C can be reassuring.)
    • “Should I continue my other medications/supplements during therapy? Any interactions?”
    • “Are there any lifestyle changes I should make to help the treatment or manage side effects?” (Diet, exercise, etc. – shows the doctor you’re proactive.)
    • “What symptoms should I report immediately?” (This helps you know what is an emergency vs what can wait for a scheduled visit.)
  • Consider a Second Opinion: If you have any doubts or just want peace of mind, it’s perfectly acceptable to seek a second opinion​. Good doctors will not be offended – in fact, they often encourage it, especially if a patient wants more reassurance. A second opinion can confirm the treatment plan or offer alternative perspectives. This is especially useful if you’re at a smaller center and can get an opinion from a major cancer center with specialists in advanced prostate cancer. Many patients do second opinions at decision points (like when first diagnosed metastatic, or when disease becomes castration-resistant) to gather more information. Insurance often covers it, but check to be sure. If travel is an issue, sometimes an inter-institution consult can be done (or telemedicine, which has grown recently).

     

  • Utilize Educational Resources: Ask your doctor for educational materials or reliable websites to learn more. They might have pamphlets on your treatment or suggest resources like Cancer Academy , Malecare , etc. But also be cautious: not all information on the internet is accurate or relevant. It’s good to run things you read by your doctor. For instance, if you read about an alternative therapy or a new drug in the news, bring it up: “I heard about XYZ treatment – is that something for me?” Your doctor can clarify if it’s real, available, or appropriate. This can clear up misconceptions and also signals to your doctor that you’re engaged in your care.

  • Practical Matters: Discuss logistical or practical concerns too. If you have trouble with transportation to frequent appointments, let them know – a social worker might arrange transport or local lab checks. If cost or insurance coverage is a concern for a certain treatment, ask to speak to a financial counselor or ask if there are assistance programs. These issues can often be solved if addressed upfront.

  • Psychosocial Support: Talk about how you’re coping. Oncologists know that advanced cancer is not just a physical challenge but an emotional one. If you feel anxious or depressed, bring it up – “This is all a lot to handle; are there support services you recommend?” They may refer you to a counselor, or a support group, or a palliative care specialist for extra support. Sometimes just having that conversation opens the door for more holistic care.

Remember, you are the most important member of your care team. Good communication with your doctors leads to better care and satisfaction. Don’t shy away from asking questions like “What’s the next step?”, “Why this treatment over another?”, “How will this affect my daily life?”. Doctors want patients to understand and be on board with the plan. As one expert advised patients: “Talk about side effects. Ask about two- and three-treatment combinations and how they will make you feel. Ask: ‘What are the benefits? What are the risks?’”​. By staying informed and engaged, you empower yourself and help your doctors help you better.

It can also be helpful to have a family member present to take notes and ask questions, as it’s easy to feel overwhelmed and miss information in the moment. And if ever in doubt, getting a second opinion is a reasonable step for your peace of mind​. Ultimately, the goal is for you and your doctor to work as a team, making decisions together based on the best evidence (which this guide has summarized) and your personal values. With open dialogue, you can ensure that you’re receiving the best possible care for your situation and that you feel comfortable and confident in the treatment plan moving forward.

 

7. Citations (References)

  1. Agarwal N, et al. JAMA Netw Open. 2022;5(12):e2249256. (Study on real-world use of combination therapy)​ cancer.gov
  2. Fizazi K, et al. N Engl J Med. 2017;377(4):352-360. (LATITUDE trial of abiraterone in mHSPC)​
  3. Davis ID, et al. N Engl J Med. 2019;381(2):121-131. (ENZAMET trial of enzalutamide in mHSPC)​

    .

  4. Chi KN, et al. N Engl J Med. 2019;381(1):13-24. (TITAN trial of apalutamide in mHSPC)​

    .

  5. Sweeney C, et al. N Engl J Med. 2015;373(8):737-746. (CHAARTED trial of docetaxel in mHSPC)​

  6. Smith MR, et al. N Engl J Med. 2022;386(12):1132-1142. (ARASENS trial of darolutamide in mHSPC)​

    .

  7. Parker C, et al. N Engl J Med. 2013;369(3):213-223. (ALSYMPCA trial of Radium-223 in mCRPC)​

    .

  8. Sartor O, et al. N Engl J Med. 2021;385(12):1091-1103. (VISION trial of Lu-177 PSMA in mCRPC)​

    .

  9. Kantoff PW, et al. N Engl J Med. 2010;363(5):411-422. (IMPACT trial of sipuleucel-T in mCRPC)​

    .

  10. de Bono J, et al. N Engl J Med. 2020;382(22):2091-2102. (PROfound trial of olaparib in mCRPC)​

  11. Karzai FH, et al. Cancer. 2021;127(10):1582-1592. (Study on AA vs White outcomes on AR therapy)​

     

  12. Agarwal N, et al. Cancer. 2021;127(24):4676-4684. (Analysis of AA vs White outcomes on docetaxel)​

    .

  13. Conibear J, et al. Clin Oncol (R Coll Radiol). 2020;32(12):757-770. (Review of PSMA-targeted therapies)​

    .

  14. Collinson F, et al. Lancet Oncol. 2021;22(2):279-290. (CARD trial of cabazitaxel vs abiraterone/enzalutamide)​

    .