Edward Weber, MD

Edward Weber, MD

Seattle, Washington

Edward Weber, MD, is a retired medical oncologist living in Seattle, Washington. He was born and raised in a suburb of Reading, Pennsylvania. After graduating from Princeton University in 1956 with a BA in History, Dr. Weber attended medical school at the University of Pennsylvania. His internship training took place at the University of Vermont in Burlington.

A tour of service as a Naval Flight Surgeon positioned him on Whidbey Island, Washington, and this introduction to the Pacific Northwest ultimately proved irresistible. Following naval service, he received postgraduate training in internal medicine in Philadelphia at the Pennsylvania Hospital and then pursued a fellowship in hematology and oncology at the University of Washington.

His career in medical oncology was at the Tumor Institute of the Swedish Hospital in Seattle where his practice focused largely on the treatment of patients experiencing lung, breast, colon, and genitourinary cancer and malignant lymphoma.

Toward the end of his career, he developed a particular concentration on the treatment of prostate cancer. Since retirement in 2002, he has authored the PCa Commentary, published by the Prostate Cancer Treatment Research Foundation, an analysis of new developments in the prostate cancer field with essays discussing and evaluating treatment management options in this disease. He is a regular speaker at various prostate cancer support groups around Seattle.

Disclosures:

Talks by Edward Weber, MD

PCa Commentary | Volume 190 – June 2024

Non-Metastatic Castration-Resistant Prostate Cancer:
Management Options, Associated Adverse Effects, and Treatment Outcomes.

‘Non-metastatic castration-resistant prostate cancer’ (nmCRPC) applies to a wide variety of patients with different risks for disease progression. It refers to patients who have had primary therapy intended for cure who then experience a rising PSA while receiving initial hormone suppression, i.e., nmCRPC. The ‘non-metastatic’ status designates that despite a rising PSA while on ADT conventional imaging with CT and bone scan is negative for metastatic spread. Among this ‘non-metastatic‘ category are men who have features likely associated with early development of metastatic disease i.e., PSA doubling time less than or equal to 10 months and Gleason score greater than or equal to 8. A treatment goal for this group would be to delay the occurrence of metastases, i.e., prolong metastases-free survival.

Non-metastatic castration-resistant prostate cancer in men with high-risk features is frequently metastatic on PSMA PET/CT scans.

This high-risk group was studied by Fendler et al, as reported in Clin Cancer Res. 2019. The study group comprised 200 men with high-risk nmCRPC who exhibited a rising PSA while on hormone suppression (ADT): i.e., PSA greater than 2 ng/ml., PSA doubling time in less than or equal to 10 months and Gleason score greater than or equal to 8. All were negative for metastatic disease on conventional CT and bone scans. When studied with PSMA PET scans, pelvic disease was found in 44%, of which 24% had recurrence in the prostate bed and 55% had cancer spread beyond the pelvis: extra-pelvic nodes, 39%; bone, 24% and viscera disease, 6%.

Treatment Options for Non-Metastatic CRPC:

1) Continue therapy with ADT until conventional imaging detects metastases and then augment ADT with second generation androgen receptor inhibitors (SGARi) – apalutamide, enzalutamide, darolutamide or the steroidogenesis inhibitor abiraterone or initiate a SGARi if the PSA doubling time is less than months.
2) Perform a PSMA PET scan and, upon detecting metastases, augment ADT with a SGARi. The establishment of metastatic CRPC opens additional therapeutic options: Provenge, Bipolar Androgen Therapy (BAT) or metastasis directed therapy (MDT).

A caveat: A thoughtful reflection on option #1 above was presented by Madan et al., Journal of Clinical Oncology, March 2024: “Restaging with Prostate-Specific Membrane Antigen Imaging in Metastatic Castration-Resistant Prostate Cancer: When Seeing More is Detrimental to Care.” They “seriously question if identifying subtle, likely subclinical changes on PSMA imaging translates into improving care” in patients with nmCRPC. Their recommendation is to base any therapeutic switch on disease progression exhibited on CT or bone scan. They note that in nmCRPC [i.e., early in the course of the disease] the PSA can rise over a duration of 17 months
without evidence of progression on conventional imaging. Madan’s position is that a premature switch of therapy based on a PSA rise, or PSMA imaged metastases, should be withheld until research data demonstrates a clinical gain, otherwise the patient is at risk for sustaining adverse side effects without a commensurate survival benefit. A comparison of PSMA and standard imaging in biochemically recurrent prostate cancer is underway in a Clinical Trial, NCT04777071.

Comparison of outcome and quality of life with systemic therapies in men with nmCRPC whose PSA doubling time is less than 10 months.

This was addressed by Shore et al. in “A multidisciplinary approach to address unmet needs in the management of patients with non-metastatic castration-resistant prostate cancer,” Prostate Cancer and Prostatic Disease, March 2024. Shore summarized the goals of treatment for nmCRPC as “delay or prevent metastatic disease and the need for additional antimetastatic treatment, maintain quality of life and prolong survival.” In short, in this high-risk group apalutamide, enzalutamide and darolutamide all increased
metastasis-free and overall survival when added to ADT. They all performed equivalently. The choice among the three is influenced by the adverse effects associated with each. Since there has been no “head-to-head” comparison, Shore’s article offers a consensus opinion based on “a panel of 10 multidisciplinary experts:”

Findings: Fatigue is a common feature of all three drugs: 32.6% for enzalutamide; 30.4%, apalutamide; 12.1% darolutamide — all greater compared to Lupron.

Other adverse effects:

apalutamide (Erleda) – rash, 23.8%; falls 15.6% seizures, 0.2%
darolutamide (Nubeqa) – back pain, 8.8%; arthralgia, 8.1%; seizures, 0.2%
enzalutamide  (Xtandi) – hot flashes, 13%; hypertension, 11.9%; falls, 11.4% and nausea, 11.4%. Seizure rate is comparably low.
Their comparison found “a stable long-term safety profile for darolutamide, whereas the likelihood… for serious adverse effects increased with apalutamide and enzalutamide.” Discontinuation of treatment was least likely with darolutamide, 33 months as compared to enzalutamide and apalutamide, 20.8 and 18.5 months, respectively. (George et al., JCO, 2023).

Drug-drug interactions are of significance and common in the older population commonly taking these drugs. The listing in the article is extensive. A consultation with a pharmacist is appropriate to evaluate and advise regarding a patient’s potential adverse drug-drug interactions.

To be noted: The high-risk for recurrence in the men studied in Fendler (above) and evaluated by Shore (also above) share similar high-risk features, suggesting that the ‘non-metastatic’ patients in Shore, if evaluated by PET/CT, would most likely be PET positive for metastatic CRPC.

A Counterintuitive relationship:  CT progression associated with a non-rising PSA in late disease.

Encompassed in the Shore article is the statement: “ … a substantial proportion of patients using SGARis may experience disease progression without rising PSA levels.”  This counterintuitive finding was reported initially and analyzed in 2017 in “Radiographic progression with non-rising PSA in metastatic castration-resistant prostate cancer: a post hoc analysis of [the] PREVAIL [trial]” (Bryce et al, Prostate Cancer and Prostatic Diseases, 2017). The PREVAIL trial compared enzalutamide to inert placebo pills in men with metastatic progression after initial hormone therapy as diagnosed by CT, bone scan and MRI. Bryce reported that “among 265
patients with radiographic progression, 65 (24.5%) had non-rising PSA levels.”

In their study, one-third of men with non-rising PSA, but with progression on imaging, had visceral disease, a common feature in men with neuroendocrine prostate cancer, either mixed with adenocarcinoma or pure NEPC. Whereas PSMA PET scans do not register NEPC because of the lack of the PSMA target, in CT scans the physical presence of lesions containing NEPC are
imaged.

With available data in 2017, Bryce et al. could only speculate that CT progression without PSA increase resulted from an admixture of adenocarcinoma and NEPC in which the NEPC lesions were CT apparent but not secreting PSA. In the future when the epigenetic test, “NEMO”, devised by Bartran (discussed in Commentary #187) becomes commercially available the extent of NEPC in this mixed situation can be assessed on circulating tumor DNA.

BOTTOM LINE:
Men with ‘non-metastatic’ CRPC at high risk for recurrence, when scanned with a PSMA PET, will be found to have metastatic CRPC in nearly all cases. Men designated as ‘nmCRPC’ and those men established as having metastases, are equally responsive to the addition of second-generation androgen receptor inhibitors when added to ongoing ADT.

Read More

PCa Commentary | Volume 189 – May 2024

Non-Metastatic Castration-Resistant Prostate Cancer:
Management Options, Associated Adverse Effects, and Treatment Outcomes.

‘Non-metastatic castration-resistant prostate cancer’ (nmCRPC) applies to a wide variety of patients with different risks for disease progression. It refers to patients who have had primary therapy intended for cure who then experience a rising PSA while receiving initial hormone suppression, i.e., nmCRPC. The ‘non-metastatic’ status designates that despite a rising PSA while on ADT conventional imaging with CT and bone scan is negative for metastatic spread. Among this ‘non-metastatic‘ category are men who have features likely associated with early development of metastatic disease i.e., PSA doubling time less than or equal to 10 months and Gleason score greater than or equal to 8. A treatment goal for this group would be to delay the occurrence of metastases, i.e., prolong metastases-free survival.

Non-metastatic castration-resistant prostate cancer in men with high-risk features is frequently metastatic on PSMA PET/CT scans.

This high-risk group was studied by Fendler et al, as reported in Clin Cancer Res. 2019. The study group comprised 200 men with high-risk nmCRPC who exhibited a rising PSA while on hormone suppression (ADT): i.e., PSA greater than 2 ng/ml., PSA doubling time in less than or equal to 10 months and Gleason score greater than or equal to 8. All were negative for metastatic disease on conventional CT and bone scans. When studied with PSMA PET scans, pelvic disease was found in 44%, of which 24% had recurrence in the prostate bed and 55% had cancer spread beyond the pelvis: extra-pelvic nodes, 39%; bone, 24% and viscera disease, 6%.

Treatment Options for Non-Metastatic CRPC:

1) Continue therapy with ADT until conventional imaging detects metastases and then augment ADT with second generation androgen receptor inhibitors (SGARi) – apalutamide, enzalutamide, darolutamide or the steroidogenesis inhibitor abiraterone or initiate a SGARi if the PSA doubling time is less than months.
2) Perform a PSMA PET scan and, upon detecting metastases, augment ADT with a SGARi. The establishment of metastatic CRPC opens additional therapeutic options: Provenge, Bipolar Androgen Therapy (BAT) or metastasis directed therapy (MDT).

A caveat: A thoughtful reflection on option #1 above was presented by Madan et al., Journal of Clinical Oncology, March 2024: “Restaging with Prostate-Specific Membrane Antigen Imaging in Metastatic Castration-Resistant Prostate Cancer: When Seeing More is Detrimental to Care.” They “seriously question if identifying subtle, likely subclinical changes on PSMA imaging translates into improving care” in patients with nmCRPC. Their recommendation is to base any therapeutic switch on disease progression exhibited on CT or bone scan. They note that in nmCRPC [i.e., early in the course of the disease] the PSA can rise over a duration of 17 months
without evidence of progression on conventional imaging. Madan’s position is that a premature switch of therapy based on a PSA rise, or PSMA imaged metastases, should be withheld until research data demonstrates a clinical gain, otherwise the patient is at risk for sustaining adverse side effects without a commensurate survival benefit. A comparison of PSMA and standard imaging in biochemically recurrent prostate cancer is underway in a Clinical Trial, NCT04777071.

Comparison of outcome and quality of life with systemic therapies in men with nmCRPC whose PSA doubling time is less than 10 months.

This was addressed by Shore et al. in “A multidisciplinary approach to address unmet needs in the management of patients with non-metastatic castration-resistant prostate cancer,” Prostate Cancer and Prostatic Disease, March 2024. Shore summarized the goals of treatment for nmCRPC as “delay or prevent metastatic disease and the need for additional antimetastatic treatment, maintain quality of life and prolong survival.” In short, in this high-risk group apalutamide, enzalutamide and darolutamide all increased
metastasis-free and overall survival when added to ADT. They all performed equivalently. The choice among the three is influenced by the adverse effects associated with each. Since there has been no “head-to-head” comparison, Shore’s article offers a consensus opinion based on “a panel of 10 multidisciplinary experts:”

Findings: Fatigue is a common feature of all three drugs: 32.6% for enzalutamide; 30.4%, apalutamide; 12.1% darolutamide — all greater compared to Lupron.

Other adverse effects:

apalutamide (Erleda) – rash, 23.8%; falls 15.6% seizures, 0.2%
darolutamide (Nubeqa) – back pain, 8.8%; arthralgia, 8.1%; seizures, 0.2%
enzalutamide  (Xtandi) – hot flashes, 13%; hypertension, 11.9%; falls, 11.4% and nausea, 11.4%. Seizure rate is comparably low.
Their comparison found “a stable long-term safety profile for darolutamide, whereas the likelihood… for serious adverse effects increased with apalutamide and enzalutamide.” Discontinuation of treatment was least likely with darolutamide, 33 months as compared to enzalutamide and apalutamide, 20.8 and 18.5 months, respectively. (George et al., JCO, 2023).

Drug-drug interactions are of significance and common in the older population commonly taking these drugs. The listing in the article is extensive. A consultation with a pharmacist is appropriate to evaluate and advise regarding a patient’s potential adverse drug-drug interactions.

To be noted: The high-risk for recurrence in the men studied in Fendler (above) and evaluated by Shore (also above) share similar high-risk features, suggesting that the ‘non-metastatic’ patients in Shore, if evaluated by PET/CT, would most likely be PET positive for metastatic CRPC.

A Counterintuitive relationship:  CT progression associated with a non-rising PSA in late disease.

Encompassed in the Shore article is the statement: “ … a substantial proportion of patients using SGARis may experience disease progression without rising PSA levels.”  This counterintuitive finding was reported initially and analyzed in 2017 in “Radiographic progression with non-rising PSA in metastatic castration-resistant prostate cancer: a post hoc analysis of [the] PREVAIL [trial]” (Bryce et al, Prostate Cancer and Prostatic Diseases, 2017). The PREVAIL trial compared enzalutamide to inert placebo pills in men with metastatic progression after initial hormone therapy as diagnosed by CT, bone scan and MRI. Bryce reported that “among 265
patients with radiographic progression, 65 (24.5%) had non-rising PSA levels.”

In their study, one-third of men with non-rising PSA, but with progression on imaging, had visceral disease, a common feature in men with neuroendocrine prostate cancer, either mixed with adenocarcinoma or pure NEPC. Whereas PSMA PET scans do not register NEPC because of the lack of the PSMA target, in CT scans the physical presence of lesions containing NEPC are
imaged.

With available data in 2017, Bryce et al. could only speculate that CT progression without PSA increase resulted from an admixture of adenocarcinoma and NEPC in which the NEPC lesions were CT apparent but not secreting PSA. In the future when the epigenetic test, “NEMO”, devised by Bartran (discussed in Commentary #187) becomes commercially available the extent of NEPC in this mixed situation can be assessed on circulating tumor DNA.

BOTTOM LINE:
Men with ‘non-metastatic’ CRPC at high risk for recurrence, when scanned with a PSMA PET, will be found to have metastatic CRPC in nearly all cases. Men designated as ‘nmCRPC’ and those men established as having metastases, are equally responsive to the addition of second-generation androgen receptor inhibitors when added to ongoing ADT.

Read More

PCa Commentary | Volume 188 – April 2024

Prostate cancer is heterogeneous throughout  –  within the prostate are divergent clones of varying malignant potential vying for escape. Within and among metastatic sites heterogeneity reigns. This complex biology presents diagnostic challenges and complicates management decisions. This Commentary reviews a seminal article which comprehensively researched this issue and provides clinical insight.

“Reversible epigenetic alterations mediate PSMA expression heterogeneity in advanced metastatic prostate cancer,” Sayer, Haffner and 22 colleagues, JCI insight, Feb 2023.

The key to their findings is the powerful tool of “rapid autopsy” wherein the prostate and multiple metastases sites are extensively biopsied directly after a patient’s death. Although PSMA is extensively expressed in localize cancer, “imaging studies of metastatic castration-resistant prostate cancer have demonstrated that up to 30% of patients have PSMA-negative tumors.”  Of 52 men (339 sites evaluated) with metastatic disease 25% had no detectable PSMA at any site. (Prostate Specific Membrane Antigen is the cell surface protein found in varying intensity on prostate cells). 44% of men “showed heterogeneous PSMA expression across individual metastases”. A single lesion might have both high and low PSMA expressing cells with “33 (63%) cases harboring at least one PSMA negative site.” Some cells lacking PSMA expression exhibited a biomarker for neuroendocrine transformation (NE). A small percentage of cells (7%) lacked expression of both PSMA and neuroendocrine markers. This heterogeneity limits the diagnostic effectiveness of PSMA PET scans in imaging the totality of a patient’s metastatic disease burden and has consequences relating to radioligand targeting of PSMA with, i.e., 177-Lutetium-PSMA-617.

Additional complexity regarding PSMA expression was seen: “Notably, we observed focal PSMA staining of the tumor-associated vasculature in tumors lacking PSMA expression in tumor cells.”

The PSMA negative tumors inversely expressed an alternative surface antigen, MUC1 (aka CEACAM5), a neuroendocrine marker whose function is to suppress a cell’s differentiation toward maturity and promote tumor growth. Varying PSMA expression was seen in different organs with lower expression in liver lesions, higher levels in adrenal glands, as compared to middling levels in the prostate and bone metastases suggesting “an interplay between the tumor microenvironment of metastatic sites and PSMA expression.”

The Sayer research discovered a therapeutically relevant relationship: “Epigenetic therapies can restore and augment PSMA expression.” They noted that a suppressive tag (epigenetic methylation) on DNA at portions of the FOLH1 gene suppressed PSMA expression and that the available drug, vorinostat (Zolinza), a histone deacetylase inhibitor, can remove this tag and partially restore PSMA expression. The effect of this agent in reversing PSMA suppression was confirmed in experiments in mice. Taken all together, the combined use of this drug with radioligand targeting of PSMA with, i.e., 177-Lu-PSMA-617 (Pluvicto), might increase the effectiveness of this regimen. The mixture of PSMA and CEACAM5 among metastases suggested a regimen of “co-targeting” both antigens with appropriate radioligands.
PCa Commentary

What are the major take-away points from this elegant research?

Heterogeneity of clones within the prostate and among metastatic sites dilute the accuracy of single site biopsies to capture the dominant genomic character of the cancer on which risk assessment and treatment selection are made. Assaying circulating free tumor DNA or sequencing pooled biopsies can provide broader coverage. 
Response to radioligand therapy with 177-Lu-PSMA-617 (Pluvicto) is proportional to the intensity of PSMA expression in the prostate and metastatic sites, as imaged by PET scans with Pylarify and Ga-68-PSMA-11. A substantial number of metastatic sites show weak PSMA expression and 25% show no PSMA expression and would be expected to respond poorly or not at all to radioligand therapy.
In the Vision trial of 177-Lu-PSMA-617 in men with advanced metastatic prostate cancer only approximately 50% of men showed a >50% decline in PSA, an outcome explained by the heterogeneity of PSMA expression among metastases. The extent of heterogeneity of PSMA expression at earlier stages of disease has not been reported.
The extent of PSMA expression in metastatic lesions is inversely proportional to the expression of markers for neuroendocrine transformation, CEACAM5 / MUC1, suggesting applicability of co-targeting of PSMA and CEACAM5 / MUC1, when radioligands for the latter are effectively developed. MUC1 can be targeted with CAR-T and antibody-drug conjugate therapies.
Epigenetically silenced PSMA expression at the FOLH1 gene can be reversed with vorinostat, a histone deacetylase inhibitor, suggesting that the effectiveness of radioligand targeting of PSMA could be enhanced by combining radioligand therapy with vorinostat.
BOTTOM LINE:
Advanced research documenting the heterogeneity of PSMA and CEACAM5 expression in metastatic prostate cancer carries significant implications regarding the diagnostic and therapeutic effectiveness of PSMA targeting in advanced disease. A regimen for restoring decreased PSMA expression is presented.

Read More

PCa Commentary | Volume 186 – February 2024

ACTIVE SURVEILLANCE: Patient Selection, Outcome and Monitoring for Gleason Grade Progression.
Question: Why not be treated at initial diagnosis of prostate cancer— and hope for cure?
Answer: Because all treatments are associated with unwelcome adverse effects that most men would prefer to avoid. Who should receive immediate treatment, and which men may safely delay treatment, preserving quality of life, — and with careful monitoring and timely intervention experience a similar outcome as if treated initially. That is the subject of this Commentary: patient selection for active surveillance (AS) and new techniques for monitoring for progression during AS.

Currently, eligibility for AS is based on clinical/pathological and biomarker features that define low- or favorable intermediate-risk prostate cancer: Gleason score 3+3 (Grade Group1) and Gleason score 3+4 (Grade Group 2); < 20% Gleason pattern 4; less than 50% positive biopsy cores and having only one NCCN intermediate risk factor (i.e., PSA 10-20 ng/ml, Gleason score 7 and cancer limited to the prostate). A PSA Density of <.15 and an MRI PIRAD score of 1 or 2 support AS. Although Gleason Grade Group 1 is to a small extent heterogeneous, the behavioral heterogeneity of Gleason score 7 grouping has led to a sub-classification into “favorable (Gleason 3+4; Gleason Grade Group 2) and unfavorable intermediate-risk cancer (4+3; Gleason Grade Group 3), the latter not advised for AS. The concern regarding the extent of Gleason pattern 4 in Gleason score 3+4 is based on the understanding that prostate cancer cells with pattern 4 characteristics have the potential to invade and metastasize. Patients with <5% pattern 4, are deemed satisfactory for AS, whereas a rise toward 20% increases the advisability for early intervention. The NCCN guidelines “prefer” AS as opposed to initial treatment for low-risk patients and allows consideration of AS for men with favorable intermediate-risk cancer with low PSA density (< .15) and low tumor volume ( i.e., < 2 positive cores), low genomic risk score and low percentage of Gleason pattern 4, i.e., <5%. Brief Summary of Outcome of Trials of Active Surveillance in Patients with Gleason Grade Groups 1 and 2: An extensive current review (Mukherjee et al., Journal Clinical Medicine, Dec. 2023) of outcomes for men with localized cancer (low-risk and favorable intermediate-risk) was based on a review of 712 studies from which 25 provided sufficient detail. Two representative studies will be briefly summarized: Courtney et al., (J.NCCN. 2022): Men on AS [8726 low-risk (LRPC) and 773 favorable intermediate-risk (FIRPC)] patients were followed for a median of 7.6 years. Metastasis-free survival at 10 years was 98.5% vs 90.4%; cancer-specific survival was 99% vs 96%, respectively. Mukhergee et al., (Eur. Urol. Open Sci., 2023): For men on AS (276 LRPC and 96 FIRPC) with median follow-up of 4.5 years. “… there was no significant difference in the median duration of AS between the two groups (32.5 months for IRPCvs 36 months for LRPC, p=0.53.” During the course of AS 30% had disease progression and were offered active treatment. The overall survival probability at 5 years for LRPC and FIRPC was 93% for both, and at 10-years 90% vs 83%s respectively. Studies from John Hopkins and Toronto report 98-99% cancer-specific survival in carefully selected and monitored men with low- and favorable intermediate-risk cancer despite 36% to 50% conversion to treatment during AS due to Gleason grade progression (Data from NCCN 2022 guidelines). The excellent survival figures in all these studies point to the effectiveness of treatment in those men who progressed during AS. The equivalence of outcome at intervention for carefully selected and monitored men on AS compared to those men treated with surgery at diagnosis has been multiply reported. Epstein, Carter et al., (Journal Urology, 2017) reported, ”Patients on active surveillance reclassified to grade group 2 or greater are at no greater risk for treatment failure than men newly diagnosed with similar grades.” Genomic Classifiers (Decipher, Prolaris, OncotypeDx) provide greater prognostic accuracy than standard clinical/pathological classifications (cited above) for estimating progression in men with localized prostate cancer considering AS. At the 2023 meeting the Society of Urologic Oncology Sheng et al. (Abst 237) reported that in a study of 235 men, Decipher Genomic Classifier (range of increasing concern for metastases and mortality extend from 0 -1.0) was associated with an upgrade to adverse pathology in men with a scores above 0.4 (p=.002) and above 0.6 (p=.006) - both values not suitable for men considering AS. A second study by Khandaker based on data from the Miami Active Surveillance Trial reported similar findings: Decipher scores >0.4 and increasingly above 0.6 were associated with adverse Gleason grade progression which would signal early intervention as opposed to AS.

The Next Step Forward: Predicting Future Progression Dynamically During AS as Opposed to Predictions Made Only at Baseline.

The clinical/pathological classification systems cited above offer prognostic (as opposed the predictive) information to guide patient selection but are not patient-specific. For example, a Decipher score of, say, 4.3 establishes a concerning level of risk but leaves the patient and physician a significant management decision about how to incorporate that extent of risk in a management plan. Studies using statistical analysis (see Cooperberg below) and artificial Intelligence (see Lee and Nayan below) provide dynamic patient-specific predictions of adverse grade progression during the course of AS.

Cooperberg et al.,(JAMA Oncol, Aug 2020) addressed this issue in “Tailoring Intensity of Active Surveillance for Low-risk Cancer Based on Individualizing Prediction of Risk Stability.”
The Canary Prostate Active Surveillance Study (PASS) involves 9 academic medical centers and based their study on 850 very explicitly followed patients for at least 5 years following enrollment. Their product provided an individualized prediction at the time of diagnosis or during AS of ‘non-reclassification’ at 4 years – i.e., information that might guide continued participation in AS or a switch to intervention. “The Canary model was built to be calculable at any given landmark time or event in the course of active surveillance.” Their findings suggest that “based on an individual’s risk parameters, that many men may be safely monitored with a substantially less intensive surveillance regimen.”

Two other studies employed AI to predict grade progression during AS [Lee et al. (Nature Prostate Journal, Digital Medicine, Aug 2022) and Nayan et al. (Urol Oncol. Apr 2022)]. Both provided a guideline regarding future progression. Using AI and incorporating interval monitoring data (PSA, MRI and biopsies) each study estimated an ongoing “real life” prediction at any time during AS of the risk of future Gleason grade reclassification, information that could influence the decision to withdraw from AS and switch to active intervention.

BOTTOM LINE:

Active Surveillance is the preferred management option for men with low- and favorable intermediate-risk prostate cancer and has been shown to yield excellent outcomes. Genomic classifiers are further refining patient selection. Statistical analysis and artificial intelligence provide dynamic risk assessment for grade progression during the ongoing course of AS.

Read More

PCa Commentary | Volume 185 – January 2024

Background

Oligometastatic prostate cancer (omPC) designates the status of having 3 to 5 metastatic lesions at diagnosis with an untreated primary or a similar extent of spread at recurrence after primary therapy. Metastasis directed therapy (MDT) focuses radiation to those several lesions. This situation is increasingly prevalent due to the more frequent use of PSMA PET imaging and in 2018 the incidence of hormone sensitive metastatic PC prostate cancer (mHSPC) at diagnosis was 8.2% (Vandenberg et al. Prostate Cancer and Prostatic Diseases, 2023), occurring mainly in men with high-risk cancer. Metastatic hormone sensitive PC can be found in men studied by PET imaging whose PSA is rising following primary therapy, converting non-metastatic HSPC to metastatic hormone resistant PC (mHRPC).

In their study of 200 men with rising PSA values after primary therapy before hormone intervention, PSMA PET scans identified metastases beyond the prostate in 55% of men, pelvic nodal disease in 20% and local recurrence in 24% (ibid). Studies comparing the genomics of denovo mHSPC with recurrent mHRPC have found — likely due to the delay in diagnosis allowing time for mutations to develop — that the cancer in the recurrent state is more aggressive.

Focal radiation therapy (e.g. with CyberKnife radiation) achieves >95% local control of oligometastatic lesions, but the major deficiency of MDT is the subsequent emergence of un-imaged polymetastatic disease. Metastatic prostate cancer is biologically heterogeneous with some metastatic sites remaining indolent and others rapidly progressing to polymetastatic spread. The current research challenge is to identify biologic markers and molecular signatures to predict metastatic behavior and guide therapy based on which men would benefit from MDT.

Three treatment options for oligometastatic prostate cancer were reviewed in PCa Commentary Vol. #182. In brief, MDT without ADT, MDT with ADT, and MDT combined with intermittent Xtandi. All had improved overall survival as compared to ADT only. However, in those studies patients were selected based on CT and technetium bone scans, not the more sensitive PSMA PET imaging, which detects metastases earlier at diagnosis and at recurrence. Current patient selection is based arbitrarily on the number and location of treatable metastatic lesions.

Conventional Predictors of Progression Following MDT for Oligometastatic Prostate Cancer

Analysis of early trials comparing MDT with no ADT (Deek, JCO 2022) found that men with mutations in the DNA damage repairs genes, i.e. BRCA 1, 2 and ATM, were at high risk for early failure. Radiographic progression-free survival for those without deleterious mutations compared to those with mutations was 22.6 months vs 10 months, respectively.

Another analysis found that the size, location of metastatic lesions and PSA doubling time in men with oligorecurrent disease affected outcome (Franzese, Clin & Experimental Metastases, 2022). In their study with PSMA PET imaging the median size of metastatic lesions was 4 cm.

Local control at 1 and 2 years was 94% and 92%; progression-free survival at 1 and 2 years, 80% and 69%, with a median time to progression of 33.7 months. The best outcomes were associated with pelvic nodal disease, followed by extra-pelvic nodal spread or metastases to bone. The take-away from these data is the need for more accurate predictors based on molecular features to guide selection of men who will benefit most from MDT.

Liquid Biopsy

The current quest is to identify a molecular signature to predict the aggressiveness of oligometastatic lesions to determine the likelihood of rapid progression to polymetastatic disease.

Tumor cells circulate (CTC) in the blood pre-diagnosis and increasingly as the disease progresses. Cell Search, an FDA approved test, has established that 5 or more CTC in 7.5 cc of blood augers a poor prognosis and less than 4 is associated with a better prognosis. Sophisticated genomic sequencing – usually referred to as ‘next generation sequencing,’ can analyze the DNA and identify the associated mutations of these circulating cells. The analysis of DNA debris from these cells – referred to as circulating tumor DNA (ctDNA) summarizes the contribution of ctDNA of the entire malignant population, an advantage since the genomes of various metastatic lesions may differ, rendering a biopsy of one site a limited representation of the overall metastatic burden.

Most circulating free DNA is shed from normal cells; in cancer patients only 0.01 – 5% of the circulating free DNA is derived from tumor cells. After the primary prostate tumor has been removed or treated, ctDNA characterizes the totality of micro- or macro metastases – and it is this ctDNA that is being evaluated for clues as to the aggressiveness of the oligometastatic lesions to guide the appropriateness of MDT.

A Major Effort

“Stratification of Oligometastatic Prostate Cancer by Liquid Biopsy: Clinical Insights from a Pilot Study,” Colosini, Triggiano et al, Biomedicines, 2022. In their earlier abstract (GU Cancers Symposium, 2018) they described the background of their study: “oligometastatic prostate cancer (OPC) may represent the initial stage of an unfavorable, rapid progression to a polymetastatic state, or the expression of a real oligometastatic phenotype related to a condition of stable disease for a long time.” They studied 28 men with hormone naive OPC, imaged with 11C-Choline PET CT, sequencing a panel of 37 prostate cancer relevant genes in circulating free DNA and microRNA. The genomic analysis was repeated frequently to evaluate the evolution to disease progression. The commonest adverse mutations were ATM, 50%, BRCA2, 39% and BRCA1 21%.

Having established the pre-MDT genomic characteristics of the men, their goal is in the subset follow-up to relate the molecular biomarker to outcome. The study is immature for analysis, but it is hoped that the outcome will provide guidance for patient selection to MDT.

BOTTOM LINE

Metastasis directed therapy prolongs survival in men with oligometastatic prostate cancer. It is hoped that genomic analysis with liquid biopsy will provide guidance for improved patient selection.

Read More

Join the GRU Community

- Why Join? -