1
|
Romero Crespo A, Carrasco Gomez D, Sanchez Martinez NM, Bautista Vidal C, Flores Sirvent L, España Navarro R, Garcia Galisteo E. Prostate-specific antigen doubling time in prostate cancer: a review of calculation methods and clinical implications. Curr Opin Urol 2025:00042307-990000000-00254. [PMID: 40371530 DOI: 10.1097/mou.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
PURPOSE OF REVIEW Prostate-specific antigen doubling time (PSADT) is a key prognostic marker in prostate cancer, especially in cases of biochemical recurrence. It guides risk stratification and therapeutic decisions, but its calculation varies significantly across institutions. This review addresses the clinical relevance of PSADT and the need for standardized methods. RECENT FINDINGS Multiple models exist for PSADT calculation, including logarithmic regression, linear models, and nadir subtraction. Logarithmic regression best reflects PSA's exponential growth but requires at least three PSA values over time. Measurement variability - due to assay differences or sampling intervals - can affect PSADT estimates by 15-40%, impacting treatment decisions. Correction tools like the Memorial Sloan Kettering Cancer Center's Excel calculator have reduced variability by up to 30-40%, improving reliability. SUMMARY PSADT remains a valuable tool in prostate cancer management, particularly for guiding salvage therapies in biochemical recurrence. However, its accuracy depends on the calculation method and PSA measurement quality. Integrating validated tools like the MSKCC calculator and adopting standardized approaches can enhance clinical decision-making. This review underscores the need for improved harmonization of PSADT methodology across guidelines and clinical settings.
Collapse
Affiliation(s)
- Alfonso Romero Crespo
- Hospital Regional Universitario de Málaga: Hospital Regional Universitario de Malaga, Malaga, Spain
| | | | | | | | | | | | | |
Collapse
|
2
|
Rasmussen KM, Patil V, Li C, Yong C, Appukkutan S, Grossman JP, Jhaveri J, Halwani AS. Survival Outcomes by Race and Ethnicity in Veterans With Nonmetastatic Castration-Resistant Prostate Cancer. JAMA Netw Open 2023; 6:e2337272. [PMID: 37819658 PMCID: PMC10568364 DOI: 10.1001/jamanetworkopen.2023.37272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/28/2023] [Indexed: 10/13/2023] Open
Abstract
Importance Racial and ethnic disparities in prostate cancer are poorly understood. A given disparity-related factor may affect outcomes differently at each point along the highly variable trajectory of the disease. Objective To examine clinical outcomes by race and ethnicity in patients with nonmetastatic castration-resistant prostate cancer (nmCRPC) within the US Veterans Health Administration. Design, Setting, and Participants A retrospective, observational cohort study using electronic health care records (January 1, 2006, to December 31, 2021) in a nationwide equal-access health care system was conducted. Mean (SD) follow-up time was 4.3 (3.3) years. Patients included in the analysis were diagnosed with prostate cancer from January 1, 2006, to December 30, 2020, that progressed to nmCRPC defined by (1) increasing prostate-specific antigen levels, (2) ongoing androgen deprivation, and (3) no evidence of metastatic disease. Patients with metastatic disease or death within the landmark period (3 months after the first nmCRPC evidence) were excluded. Main Outcomes and Measures The primary outcome was time from the landmark period to death or metastasis; the secondary outcome was overall survival. A multivariate Cox proportional hazards model, Kaplan-Meier estimates, and adjusted survival curves were used to evaluate outcome differences by race and ethnicity. Results Of 12 992 patients in the cohort, 826 patients identified as Hispanic (6%), 3671 as non-Hispanic Black (28%; henceforth Black), 7323 as non-Hispanic White (56%; henceforth White), and 1172 of other race and ethnicity (9%; henceforth other, including American Indian or Alaska Native, Asian, Native Hawaiian or Other Pacific Islander, unknown by patient, and patient declined to answer). Median time elapsed from nmCRPC to metastasis or death was 5.96 (95% CI, 5.58-6.34) years for Black patients, 5.62 (95% CI, 5.11-6.67) years for Hispanic patients, 4.11 (95% CI, 3.96-4.25) years for White patients, and 3.59 (95% CI, 3.23-3.97) years for other patients. Median unadjusted overall survival was 6.26 (95% CI, 6.03-6.46) years among all patients, 8.36 (95% CI, 8.0-8.8) years for Black patients, 8.56 (95% CI, 7.3-9.7) years for Hispanic patients, 5.48 (95% CI, 5.2-5.7) years for White patients, and 4.48 (95% CI, 4.1-5.0) years for other patients. Conclusions and Relevance The findings of this cohort study of patients with nmCRPC suggest that differences in outcomes by race and ethnicity exist; in addition, Black and Hispanic men may have considerably improved outcomes when treated in an equal-access setting.
Collapse
Affiliation(s)
- Kelli M. Rasmussen
- University of Utah School of Medicine, Salt Lake City
- George E. Wahlen Veterans Health Administration, Salt Lake City, Utah
| | - Vikas Patil
- University of Utah School of Medicine, Salt Lake City
- George E. Wahlen Veterans Health Administration, Salt Lake City, Utah
| | - Chunyang Li
- University of Utah School of Medicine, Salt Lake City
- George E. Wahlen Veterans Health Administration, Salt Lake City, Utah
| | - Christina Yong
- University of Utah School of Medicine, Salt Lake City
- George E. Wahlen Veterans Health Administration, Salt Lake City, Utah
| | | | | | | | - Ahmad S. Halwani
- University of Utah School of Medicine, Salt Lake City
- George E. Wahlen Veterans Health Administration, Salt Lake City, Utah
- Huntsman Cancer Institute, Salt Lake City, Utah
| |
Collapse
|
3
|
Keizman D, Frenkel M, Peer A, Kushnir I, Rosenbaum E, Sarid D, Leibovitch I, Mano R, Yossepowitch O, Margel D, Wolf I, Geva R, Dresler H, Rouvinov K, Rapoport N, Eliaz I. Modified Citrus Pectin Treatment in Non-Metastatic Biochemically Relapsed Prostate Cancer: Results of a Prospective Phase II Study. Nutrients 2021; 13:nu13124295. [PMID: 34959847 PMCID: PMC8706421 DOI: 10.3390/nu13124295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Optimal therapy of biochemically relapsed prostate cancer (BRPC) after local treatment is elusive. An established modified citrus pectin (PectaSol®, P-MCP), a dietary polysaccharide, is an established antagonist of galectin-3, a carbohydrate-binding protein involved in cancer pathogenesis. Based on PSA dynamics, we report on the safety and the primary outcome analysis of a prospective phase II study of P-MCP in non-metastatic BRPC based. Sixty patients were enrolled, and one patient withdrew after a month. Patients (n = 59) were given P-MCP, 4.8 grams X 3/day, for six months. The primary endpoint was the rate without PSA progression and improved PSA doubling time (PSADT). Secondary endpoints were the rate without radiologic progression and toxicity. Patients that did not progress by PSA and radiologically at six months continued for an additional twelve months. After six months, 78% (n = 46) responded to therapy, with a decreased/stable PSA in 58% (n = 34), or improvement of PSADT in 75% (n = 44), and with negative scans, and entered the second twelve months treatment phase. Median PSADT improved significantly (p = 0.003). Disease progression during the first 6 months was noted in only 22% (n = 13), with PSA progression in 17% (n = 10), and PSA and radiologic progression in 5% (n = 3). No patients developed grade 3 or 4 toxicity.
Collapse
Affiliation(s)
- Daniel Keizman
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
- Correspondence:
| | - Moshe Frenkel
- Department of Oncology, Rambam Medical Center, Haifa 3109601, Israel; (M.F.); (A.P.)
| | - Avivit Peer
- Department of Oncology, Rambam Medical Center, Haifa 3109601, Israel; (M.F.); (A.P.)
| | - Igal Kushnir
- Department of Oncology, Meir Medical Center and Sackler School of Medicine, Tel-Aviv University, Kfar-Saba 4428164, Israel; (I.K.); (N.R.)
| | - Eli Rosenbaum
- Department of Oncology, Rabin Medical Center, Petah-Tikva 4941492, Israel;
| | - David Sarid
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ilan Leibovitch
- Department of Urology, Meir Medical Center, Kfar-Saba 4439246, Israel;
| | - Roy Mano
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 69978, Israel; (R.M.); (O.Y.)
| | - Ofer Yossepowitch
- Department of Urology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 69978, Israel; (R.M.); (O.Y.)
| | - David Margel
- Department of Urology, Rabin Medical Center, Petah-Tikva 4941492, Israel;
| | - Ido Wolf
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Ravit Geva
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (D.S.); (I.W.); (R.G.)
| | - Hadas Dresler
- Department of Oncology, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
| | - Keren Rouvinov
- Department of Oncology, Soroka Medical Center, Beer-Sheva 8428760, Israel;
| | - Noa Rapoport
- Department of Oncology, Meir Medical Center and Sackler School of Medicine, Tel-Aviv University, Kfar-Saba 4428164, Israel; (I.K.); (N.R.)
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA 95401, USA;
| |
Collapse
|
4
|
Pachynski RK, Morishima C, Szmulewitz R, Harshman L, Appleman L, Monk P, Bitting RL, Kucuk O, Millard F, Seigne JD, Fling SP, Maecker HT, Duault C, Ramchurren N, Hess B, D'Amico L, Lacroix A, Kaiser JC, Morre M, Grégoire A, Cheever M, Yu EY, Fong L. IL-7 expands lymphocyte populations and enhances immune responses to sipuleucel-T in patients with metastatic castration-resistant prostate cancer (mCRPC). J Immunother Cancer 2021; 9:e002903. [PMID: 34452927 PMCID: PMC8404457 DOI: 10.1136/jitc-2021-002903] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Sipuleucel-T (sip-T) is a Food and Drug Administration (FDA)-approved autologous cellular immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). We hypothesized that combining sip-T with interleukin (IL)-7, a homeostatic cytokine that enhances both B and T cell development and proliferation, would augment and prolong antigen-specific immune responses against both PA2024 (the immunogen for sip-T) and prostatic acid phosphatase (PAP). METHODS Fifty-four patients with mCRPC treated with sip-T were subsequently enrolled and randomized 1:1 into observation (n=26) or IL-7 (n=28) arms of a phase II clinical trial (NCT01881867). Recombinant human (rh) IL-7 (CYT107) was given weekly×4. Immune responses were evaluated using flow cytometry, mass cytometry (CyTOF), interferon (IFN)-γ ELISpot, 3H-thymidine incorporation, and ELISA. RESULTS Treatment with rhIL-7 was well tolerated. For the rhIL-7-treated, but not observation group, statistically significant lymphocyte subset expansion was found, with 2.3-2.6-fold increases in CD4+T, CD8+T, and CD56bright NK cells at week 6 compared with baseline. No significant differences in PA2024 or PAP-specific T cell responses measured by IFN-γ ELISpot assay were found between rhIL-7 and observation groups. However, antigen-specific T cell proliferative responses and humoral IgG and IgG/IgM responses significantly increased over time in the rhIL-7-treated group only. CyTOF analyses revealed pleiotropic effects of rhIL-7 on lymphocyte subsets, including increases in CD137 and intracellular IL-2 and IFN-γ expression. While not powered to detect clinical outcomes, we found that 31% of patients in the rhIL-7 group had prostate specific antigen (PSA) doubling times of >6 months, compared with 14% in the observation group. CONCLUSIONS Treatment with rhIL-7 led to a significant expansion of CD4+ and CD8+ T cells, and CD56bright natural killer (NK) cells compared with observation after treatment with sip-T. The rhIL-7 treatment also led to improved antigen-specific humoral and T cell proliferative responses over time as well as to increased expression of activation markers and beneficial cytokines. This is the first study to evaluate the use of rhIL-7 after sip-T in patients with mCRPC and demonstrates encouraging results for combination approaches to augment beneficial immune responses.
Collapse
Affiliation(s)
- Russell K Pachynski
- Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | - Russell Szmulewitz
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | - Lauren Harshman
- Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- *Current affiliation: Surface Oncology, Cambridge, MA, USA
| | | | - Paul Monk
- Ohio State University James Cancer Hospital, Columbus, Ohio, USA
| | | | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - John D Seigne
- Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Steven P Fling
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Caroline Duault
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | | | - Bruce Hess
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Leonard D'Amico
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | - Judith C Kaiser
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - Martin Cheever
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Evan Y Yu
- University of Washington, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Lawrence Fong
- University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
5
|
Langlais CS, Chan JM. Opportunities and challenges for research on low-carbohydrate diets in prostate cancer. Nat Rev Urol 2020; 17:437-438. [PMID: 32358563 DOI: 10.1038/s41585-020-0326-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Crystal S Langlais
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA.
| | - June M Chan
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA.,Department of Urology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
6
|
Kessler ER, Su LJ, Gao D, Torkko KC, Wacker M, Anduha M, Chronister N, Maroni P, Crawford ED, Flaig TW, Glode LM, Lam ET. Phase II Trial of Acai Juice Product in Biochemically Recurrent Prostate Cancer. Integr Cancer Ther 2018; 17:1103-1108. [PMID: 30289005 PMCID: PMC6247557 DOI: 10.1177/1534735418803755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Plant derivatives have been studied as therapies for prostate cancer based on their purported anti-inflammatory and antioxidant properties and low toxicities. The acai berry is an example of a plant rich in phytochemicals, which may slow the growth of prostate cancer. METHODS This was a phase II, Simon 2-stage clinical trial in patients with biochemically recurrent prostate cancer with a primary endpoint of prostate-specific antigen (PSA) response. Patients were asymptomatic, with a rising PSA of at least 0.2 ng/mL, and were treated with twice daily intake of Acai Juice Product until PSA progression, with a primary endpoint of PSA response. RESULTS Twenty-one patients were enrolled in the first stage of the trial. One of those patients had a PSA response within the study time period. The PSA doubling time was lengthened in 71% of patients (95% confidence interval = 48% to 89%) on the trial, and in a small number of responders, this was sustained over an extended time. CONCLUSIONS This study did not meet its primary endpoint of 50% PSA response. Nevertheless, the overall tolerability and effects on PSA stabilization warrant further exploration in a biochemically recurrent population.
Collapse
Affiliation(s)
| | - Lih-Jen Su
- 1 University of Colorado, Aurora, CO, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Lim DM, Gulati R, Aleshin-Guendel S, Gawne A, Wingate JT, Cheng HH, Etzioni R, Yu EY. Undetectable prostate-specific antigen after short-course androgen deprivation therapy for biochemically recurrent patients correlates with metastasis-free survival and prostate cancer-specific survival. Prostate 2018; 78:10.1002/pros.23666. [PMID: 29987912 PMCID: PMC6328347 DOI: 10.1002/pros.23666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/07/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND Optimal utilization of novel therapies for advanced prostate cancer is challenging without a validated surrogate efficacy endpoint. Ongoing trials are using durable undetectable prostate-specific antigen (PSA) levels as a marker of efficacy. The clinical relevance of prolonged undetectable PSA after a short course of androgen deprivation therapy (ADT) is uncertain. METHODS The University of Washington Caisis database was queried for radical prostatectomy patients who received 6-12 months of ADT after biochemical recurrence (BCR), defined as PSA ≥0.2 ng/mL and no radiographically detectable metastasis. Proportions of men with undetectable PSA 12 and 24 months after ending ADT were compared to a hypothesized 5% rate using exact binomial tests. Associations with patient and tumor characteristics were examined using logistic regression, and associations with risk of subsequent metastasis and death were evaluated by log-rank tests. RESULTS After ineligibility exclusions, 23/93 (25%; 95%CI 16-35%; P < 0.001) and 14/93 (15%; 95%CI 9-24%; P < 0.001) had undetectable PSA 12 and 24 months after ending ADT, respectively. Detectable PSA at 12 months was associated with increased risk of metastasis (P = 0.006), prostate cancer-specific death (P = 0.028), and death from any cause (P = 0.065). Being 1 year older at diagnosis was associated with a 14% (95%CI 5-24%; P = 0.006) decrease in the odds of having a detectable PSA after controlling for PSA at diagnosis, PSA doubling time, grade group, and time from initial therapy to BCR. CONCLUSIONS This single-institution retrospective analysis shows that it is not uncommon to have undetectable PSA 12 or 24 months after a short course of ADT. No baseline prognostic characteristic other than age was associated with a durable (12 month) undetectable PSA. Because a durable undetectable PSA was associated with lower risks of metastasis and prostate cancer-specific death, it may be a reasonable clinical trial endpoint.
Collapse
Affiliation(s)
- Daniel M. Lim
- Department of Medicine, Division of Oncology, University of Washington, Seattle Cancer Care Alliance, Seattle, WA
| | - Roman Gulati
- Biostatistics and Biomathematics, Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Serge Aleshin-Guendel
- Biostatistics and Biomathematics, Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Agnes Gawne
- Biostatistics and Biomathematics, Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jonathan T. Wingate
- Biostatistics and Biomathematics, Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Heather H. Cheng
- Department of Medicine, Division of Oncology, University of Washington, Seattle Cancer Care Alliance, Seattle, WA
| | - Ruth Etzioni
- Biostatistics and Biomathematics, Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Evan Y. Yu
- Department of Medicine, Division of Oncology, University of Washington, Seattle Cancer Care Alliance, Seattle, WA
| |
Collapse
|
8
|
Paller CJ, Zhou XC, Heath EI, Taplin ME, Mayer T, Stein MN, Bubley GJ, Pili R, Hudson T, Kakarla R, Abbas MM, Anders NM, Dowling D, King S, Bruns AB, Wagner WD, Drake CG, Antonarakis ES, Eisenberger MA, Denmeade SR, Rudek MA, Rosner GL, Carducci MA. Muscadine Grape Skin Extract (MPX) in Men with Biochemically Recurrent Prostate Cancer: A Randomized, Multicenter, Placebo-Controlled Clinical Trial. Clin Cancer Res 2017; 24:306-315. [PMID: 29113986 DOI: 10.1158/1078-0432.ccr-17-1100] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/21/2017] [Accepted: 11/01/2017] [Indexed: 01/18/2023]
Abstract
Purpose: MuscadinePlus (MPX), a commercial preparation of pulverized muscadine grape skin, was evaluated as a therapeutic option for men with biochemically recurrent (BCR) prostate cancer wishing to defer androgen deprivation therapy.Experimental Design: This was a 12-month, multicenter, placebo-controlled, two-dose, double-blinded trial of MPX in 125 men with BCR prostate cancer, powered to detect a PSA doubling time (PSADT) difference of 6 months (low dose) and 12 months (high dose) relative to placebo. Participants were stratified (baseline PSADT, Gleason score) and randomly assigned 1:2:2 to receive placebo, 500 mg MPX (low), or 4,000 mg MPX (high) daily. Correlates included superoxide dismutase-2 (SOD2) genotype, lipid peroxidation, and polyphenol pharmacokinetics.Results: The evaluable population included 112 patients, all treated for at least 6 months and 62% treated for 12 months. No significant difference was found in PSADT change between control and treatment arms (P = 0.81): control 0.9 months (n = 20; range, 6.7-83.1), low dose 1.5 months (n = 52; range, 10.3-87.2), high dose 0.9 months (n = 40; range, 27.3-88.1). One high-dose patient experienced objective response. No drug-related CTCAE grade 3-4 adverse events were seen. In a preplanned exploratory analysis, PSADT pre-to-post increase was significant in the 27 (26%) genotyped patients with SOD2 Alanine/Alanine genotype (rs4880 T>C polymorphism) on MPX (pooled treatment arms; 6.4 months, P = 0.02), but not in control (1.8 months, P = 0.25).Conclusions: Compared with placebo, MPX did not significantly prolong PSADT in BCR patients over two different doses. Exploratory analysis revealed a patient population with potential benefit that would require further study. Clin Cancer Res; 24(2); 306-15. ©2017 AACR.
Collapse
Affiliation(s)
- Channing J Paller
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Xian C Zhou
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Tina Mayer
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Mark N Stein
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Glenn J Bubley
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Roberto Pili
- Roswell Park Cancer Institute, New York, New York
| | | | | | | | - Nicole M Anders
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Donna Dowling
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Serina King
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ashley B Bruns
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William D Wagner
- Wake Forest University School of Medicine, Winston-Salem, North Carolina
- Muscadine Naturals, Inc., Clemmons, North Carolina
| | - Charles G Drake
- New York-Presbyterian/Columbia University Medical Center, New York, New York
| | - Emmanuel S Antonarakis
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mario A Eisenberger
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michelle A Rudek
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gary L Rosner
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Carducci
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
van Die MD, Williams SG, Emery J, Bone KM, Taylor JMG, Lusk E, Pirotta MV. A Placebo-Controlled Double-Blinded Randomized Pilot Study of Combination Phytotherapy in Biochemically Recurrent Prostate Cancer. Prostate 2017; 77:765-775. [PMID: 28181675 PMCID: PMC5444299 DOI: 10.1002/pros.23317] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Men with biochemical recurrence of prostate cancer following local therapies often use natural supplements in an attempt to delay metastases and/or avoid the need for more aggressive treatments with undesirable side-effects. While there is a growing body of research into phytotherapeutic agents in this cohort, with some promising results, as yet no definitive recommendations can be made. This pilot study was undertaken to assess the feasibility of a fully-powered study to examine the effects of this phytotherapeutic intervention (containing turmeric, resveratrol, green tea and broccoli sprouts) on PSA doubling time in men with biochemical recurrence with a moderate PSA rise rate. METHODS A double blind, randomized, placebo-controlled parallel trial was conducted with 22 men with biochemically recurrent prostate cancer and a moderate rise rate (PSA doubling time of 4-15 months and no evidence of metastases from conventional imaging methods). Patients were randomized to either the active treatment arm or placebo for 12 weeks. The primary endpoints were feasibility of study recruitment and procedures, and measurement of proposed secondary endpoints (prostate symptoms, quality of life, anxiety, and depression as measured on the EORTC QLQ-C30 and PR-25, the IPSS and HADS). Data were collected to estimate PSA-log slopes and PSA-doubling times, using a mixed model, for both the pre-intervention and post-intervention periods. RESULTS Adherence to study protocol was excellent, and the phytotherapeutic intervention was well-tolerated, with similar numbers of mild-to-moderate adverse events in the active and placebo arms. Both the intervention and data collection methods were acceptable to participants. No statistical difference between groups on clinical outcomes was expected in this pilot study. There was between-subject variation in the PSA post treatment, but on average the active treatment group experienced a non-significant increase in the log-slope of PSA (pre-treatment doubling time = 10.2 months, post-treatment doubling time = 5.5 months), and the placebo group experienced no change in the log-slope of PSA (pre-treatment doubling time = 10.8 months, post-treatment doubling time = 10.9 months). CONCLUSION The findings suggest that a fully powered study of this combination is feasible in men with biochemically recurrent prostate cancer and a moderate PSA rise rate. Prostate 77:765-775, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M Diana van Die
- Department of General Practice, University of Melbourne, Parkville, Victoria, Australia
| | - Scott G Williams
- Department of General Practice, University of Melbourne, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Jon Emery
- Department of General Practice, University of Melbourne, Parkville, Victoria, Australia
| | - Kerry M Bone
- Integria (MediHerb), Warwick, Queensland, Australia
- New York Chiropractic College, Seneca Falls, New York
| | - Jeremy M G Taylor
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Elizabeth Lusk
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Marie V Pirotta
- Department of General Practice, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
A review of pomegranate in prostate cancer. Prostate Cancer Prostatic Dis 2017; 20:265-270. [PMID: 28440320 PMCID: PMC5555799 DOI: 10.1038/pcan.2017.19] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 01/28/2017] [Accepted: 02/22/2017] [Indexed: 01/21/2023]
Abstract
Background Preclinical studies showing that pomegranate juice and its components inhibit prostate cancer led to multiple clinical trials to determine whether pomegranate products could slow the growth of prostate cancer. This review summarizes the preclinical data and discusses the results of the clinical trials. Methods Trials targeted patients on active surveillance, neoadjuvant patients, patients with biochemical recurrence (BCR) following local therapy for prostate cancer, and patients with metastatic castration-resistant prostate cancer (mCRPC). Results In the BCR patient population, early phase II trials of both pomegranate juice and extract showed significant lengthening of PSA doubling time (PSADT), and confirmed the safety of pomegranate products. While a placebo-controlled phase III trial determined that pomegranate extract did not significantly prolong PSADT in BCR patients, a preplanned subset analysis of patients with the manganese superoxide dismutase (MnSOD) AA genotype showed greater PSADT lengthening on the pomegranate extract arm. In the neoadjuvant population, a large trial demonstrated a significant increase in urolithin A and a non-significant reduction in 8-OHdG, a marker of oxidation in prostate cancer tissue, on the pomegranate arm vs. the placebo arm. In addition, a randomized clinical trial of a polyphenol-rich multi-component food supplement tablet, including 31.25% pomegranate extract, found significant slowing of PSA increase in the food supplement arm vs. placebo in men on active surveillance and those experiencing biochemical recurrence. Conclusions Pomegranate juice and extract are safe but did not significantly improve outcomes in BCR patients in a large placebo controlled trial. However a subset of BCR patients with the MnSOD AA genotype appear to respond positively to the antioxidant effects of pomegranate treatment. Phase II trials of 100% pomegranate products in neoadjuvant patients and patients with mCRPC were negative. A multi-component food supplement showed promising results in a phase II study in active surveillance and BCR patients.
Collapse
|
11
|
Paller CJ, Denmeade SR, Carducci MA. Challenges of conducting clinical trials of natural products to combat cancer. CLINICAL ADVANCES IN HEMATOLOGY & ONCOLOGY : H&O 2016; 14:447-455. [PMID: 27379814 PMCID: PMC8521639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Numerous drugs that the US Food and Drug Administration (FDA) has approved for use in cancer therapy are derived from plants, including taxanes such as paclitaxel and vinca alkaloids such as vinblastine. Dietary supplements are another category of natural products that are widely used by patients with cancer, but without the FDA-reviewed evidence of safety and efficacy--be it related to survival, palliation, symptom mitigation, and/or immune system enhancement-that is required for therapy approval. Nearly half of patients in the United States with cancer report that they started taking new dietary supplements after being given a diagnosis of cancer. Oncologists are challenged in providing advice to patients about which supplements are safe and effective to use to treat cancer or the side effects of cancer therapy, and which supplements are antagonistic to standard treatment with chemotherapy, radiation, and/or immunotherapy. Despite the large number of trials that have been launched, the FDA has not approved any dietary supplement or food to prevent cancer, halt its growth, or prevent its recurrence. In this article, we review the primary challenges faced by researchers attempting to conduct rigorous trials of natural products, including shortages of funding due to lack of patentability, manufacturing difficulties, contamination, and lack of product consistency. We also highlight the methods used by dietary supplement marketers to persuade patients that a supplement is effective (or at least safe) even without FDA approval, as well as the efforts of the US government to protect the health and safety of its citizens by ensuring that the information used to market natural products is accurate. We close with a summary of the most widely used databases of information about the safety, efficacy, and interactions of dietary supplements.
Collapse
Affiliation(s)
- Channing J Paller
- Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Samuel R Denmeade
- Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael A Carducci
- Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
12
|
van Die MD, Bone KM, Emery J, Williams SG, Pirotta MV, Paller CJ. Phytotherapeutic interventions in the management of biochemically recurrent prostate cancer: a systematic review of randomised trials. BJU Int 2016; 117 Suppl 4:17-34. [PMID: 26898239 PMCID: PMC8631186 DOI: 10.1111/bju.13361] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To evaluate the evidence from randomised trials for the efficacy and safety of phytotherapeutic interventions in the management of biochemically recurrent (BCR) prostate cancer, indicated by prostate-specific antigen (PSA) progression, numbers progressing to/time to initiation of androgen-deprivation therapy or salvage therapy. PATIENTS AND METHODS MEDLINE (Ovid), EMBASE (Ovid), AMED (Ovid), CINAHL (EBSCO) and the Cochrane Library databases were searched. Clinical trials investigating phytotherapeutic interventions as dietary supplements or dietary components, including multi-component herbal formulations, in men with BCR prostate cancer were located. Eight of nine authors contacted for further information responded. Methodological quality was assessed using the Cochrane Collaboration's risk of bias assessment tool. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement for reporting systematic reviews was followed. RESULTS Of 23 full-text articles assessed for eligibility, five met the criteria for inclusion. Two studies were placebo controlled; two were active control trials; and one a high-/low-dose trial. The interventions were administered as isolated phytochemicals (sulphoraphane), phytotherapeutic extracts [Pomi-T (pomegranate, turmeric, green tea and broccoli sprout extract), soy, lycopene, and POMx (pomegranate extract)], or plant-derived dietary items (soy and lycopene). All studies found serum PSA levels to stabilise, decrease or rise more slowly in a significant number of men, and three studies reported stabilising or lengthening of PSA-doubling time. Studies were generally of good quality, but sample sizes were predominantly small, and durations short. CONCLUSIONS High-quality studies in this area are lacking. Sulphoraphane, lycopene, soy isoflavones, POMx, and Pomi-T are safe and well tolerated. There is limited evidence that they can affect PSA dynamics. No recommendation can be made for the use of these agents in managing prostate cancer morbidity and mortality until high-quality, fully powered studies are available. Recommendations are made for improving reproducibility and translation of findings with regard to study population, study endpoints, design, and the reporting of phytotherapeutic interventions.
Collapse
Affiliation(s)
| | - Kerry M. Bone
- Integria (MediHerb), Warwick, Qld, Australia
- New York Chiropractic College, Seneca Falls, NY, USA
| | - Jon Emery
- Department of General Practice, University of Melbourne, Parkville, Vic., Australia
| | - Scott G. Williams
- Department of General Practice, University of Melbourne, Parkville, Vic., Australia
- Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia
| | - Marie V. Pirotta
- Department of General Practice, University of Melbourne, Parkville, Vic., Australia
| | - Channing J. Paller
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
13
|
Paller CJ, Rudek MA, Zhou XC, Wagner WD, Hudson TS, Anders N, Hammers HJ, Dowling D, King S, Antonarakis ES, Drake CG, Eisenberger MA, Denmeade SR, Rosner GL, Carducci MA. A phase I study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination. Prostate 2015; 75:1518-25. [PMID: 26012728 PMCID: PMC4537354 DOI: 10.1002/pros.23024] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/01/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND New therapies are being explored as therapeutic options for men with biochemically recurrent prostate cancer (BRPC) who wish to defer androgen deprivation therapy. MPX is pulverized muscadine grape (Vitis rotundifolia) skin that contains ellagic acid, quercetin, and resveratrol and demonstrates preclinical activity against prostate cancer cells in vitro. METHODS In the phase I portion of this phase I/II study, non-metastatic BRPC patients were assigned to increasing doses of MPX (Muscadine Naturals. Inc., Clemmons, NC) in cohorts of two patients, with six patients at the highest dose, using a modified continual reassessment method. Initial dose selection was based on preclinical data showing the equivalent of 500 to 4,000 mg of MPX to be safe in mouse models. The primary endpoint was the recommended phase II dosing regimen. RESULTS The cohort (n = 14, 71% Caucasian, 29% black) had a median follow-up of 19.2 (6.2-29.7) months, median age of 61 years, and median Gleason score of 7. Four patients had possibly related gastrointestinal symptoms, including grade 1 flatulence, grade 1 soft stools, and grade 1 eructation. No other related adverse events were reported and one patient reported improvement of chronic constipation. Six of 14 patients came off study for disease progression (five metastatic, one rising PSA) after exposure for a median of 15 months. One patient came off for myasthenia gravis that was unrelated to treatment. Seven patients remain on study. The lack of dose-limiting toxicities led to the selection of 4,000 mg/d as the highest dose for further study. Median within-patient PSADT increased by 5.3 months (non-significant, P = 0.17). No patients experienced a maintained decline in serum PSA from baseline. CONCLUSION These data suggest that 4,000 mg of MPX is safe, and exploratory review of a lengthening in PSADT of a median of 5.3 months supports further exploration of MPX. Both low-dose (500 mg) and high-dose (4,000 mg) MPX are being further investigated in a randomized, multicenter, placebo-controlled, dose-evaluating phase II trial.
Collapse
Affiliation(s)
- Channing J Paller
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michelle A Rudek
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Xian C Zhou
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - William D Wagner
- Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Tamaro S Hudson
- Howard University Cancer Center, Washington, District of Columbia
| | - Nicole Anders
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Hans J Hammers
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Donna Dowling
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Serina King
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Emmanuel S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Charles G Drake
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mario A Eisenberger
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Samuel R Denmeade
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Gary L Rosner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael A Carducci
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
14
|
Pantuck AJ, Pettaway CA, Dreicer R, Corman J, Katz A, Ho A, Aronson W, Clark W, Simmons G, Heber D. A randomized, double-blind, placebo-controlled study of the effects of pomegranate extract on rising PSA levels in men following primary therapy for prostate cancer. Prostate Cancer Prostatic Dis 2015; 18:242-8. [PMID: 26169045 DOI: 10.1038/pcan.2015.32] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 06/02/2015] [Accepted: 06/02/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The primary objective of this study was to compare the effects of pomegranate juice on PSA doubling times (PSADT) in subjects with rising PSA levels after primary therapy for prostate cancer. METHODS Double-blind, placebo-controlled multi-institutional study, evaluated the effects of pomegranate liquid extract on serum PSA levels. The primary end point of this study was change in serum PSADT. Additional secondary and exploratory objectives were to evaluate the safety of pomegranate juice and to determine the interaction of manganese superoxide dismutase (MnSOD) AA genotype and pomegranate treatment on PSADT. RESULTS One-hundred eighty-three eligible subjects were randomly assigned to the active and placebo groups with a ratio of 2:1 (extract N=102; placebo N=64; juice N=17). The majority of adverse events were of moderate or mild grade. Median PSADT increased from 11.1 months at baseline to 15.6 months in the placebo group (P<0.001) compared with an increase from 12.9 months at baseline to 14.5 months in the extract group (P=0.13) and an increase from 12.7 at baseline to 20.3 in the juice group (P=0.004). However, none of these changes were statistically significant between the three groups (P>0.05). Placebo AA patients experienced a 1.8 month change in median PSADT from 10.9 months at baseline to 12.7 months (P=0.22), while extract patients experienced a 12 month change in median PSADT from 13.6 at baseline to 25.6 months (P=0.03). CONCLUSIONS Compared with placebo, pomegranate extract did not significantly prolong PSADT in prostate cancer patients with rising PSA after primary therapy. A significant prolongation in PSADT was observed in both the treatment and placebo arms. Men with the MnSOD AA genotype may represent a group that is more sensitive to the antiproliferative effects of pomegranate on PSADT; however, this finding requires prospective hypothesis testing and validation.
Collapse
Affiliation(s)
- A J Pantuck
- Department of Urology, Institute of Urologic Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - C A Pettaway
- The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - R Dreicer
- Cleveland Clinic Foundation, Cleveland, OH, USA
| | - J Corman
- Virginia Mason Medical Center, Seattle, WA, USA
| | - A Katz
- Winthrop University Hospital, Garden City, NY, USA
| | - A Ho
- Winthrop University Hospital, Garden City, NY, USA
| | - W Aronson
- 1] Department of Urology, Institute of Urologic Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA [2] VA Medical Center Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - W Clark
- Alaska Clinical Research Center, Anchorage, AL, USA
| | - G Simmons
- Five Valleys Urology, Missoula, MT, USA
| | - D Heber
- 1] Department of Urology, Institute of Urologic Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA [2] Department of Medicine and Clinical Nutrition, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
15
|
Suzman DL, Zhou XC, Zahurak ML, Lin J, Antonarakis ES. Change in PSA velocity is a predictor of overall survival in men with biochemically-recurrent prostate cancer treated with nonhormonal agents: combined analysis of four phase-2 trials. Prostate Cancer Prostatic Dis 2014; 18:49-55. [PMID: 25384338 PMCID: PMC4323734 DOI: 10.1038/pcan.2014.44] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/28/2014] [Accepted: 09/23/2014] [Indexed: 11/27/2022]
Abstract
Background Multiple phase-2 trials in men with biochemically-recurrent prostate cancer (BRPC) have assessed the impact of non-hormonal agents on PSA kinetics. We have previously demonstrated that changes in PSA kinetics correlate with metastasis-free survival; however, it is unknown whether these changes also correlate with overall survival (OS). Methods We performed a combined retrospective analysis of 146 men with BRPC treated on phase-2 trials using one of four investigational drugs: lenalidomide (n=60), marimastat (n=39), ATN-224 (n=22), and imatinib (n=25). We examined factors influencing OS, including within-subject changes in PSA kinetics (PSA slope, PSA doubling time, and PSA velocity) before and 6 months after treatment initiation. Results After a median follow up of 83.1 months, 49 of 146 men had died. In univariate Cox regression analysis, two factors were associated with OS: baseline PSA velocity and change in PSA velocity on therapy. In a landmark multivariable model, stratified by study (which controlled for age, Gleason score, type of local therapy, and use of ADT prior to metastases), baseline PSA velocity and increase in PSA velocity on therapy remained independent predictors of OS. Median OS for men with an increase in PSA velocity on treatment was 115.4 months and was not reached for men with a decrease in PSA velocity (HR=0.47, 95% CI 0.25 to 0.88; P=0.02). Conclusions This hypothesis-generating study suggests that within-subject changes in PSA velocity after initiation of non-hormonal therapy may correlate with OS in men with BRPC. If validated in prospective trials, change in PSA velocity may represent a reasonable intermediate endpoint for screening new agents in these patients.
Collapse
Affiliation(s)
- D L Suzman
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - X C Zhou
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - M L Zahurak
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - J Lin
- Kimmel Cancer Center at Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - E S Antonarakis
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| |
Collapse
|
16
|
Lin J, Zhan T, Duffy D, Hoffman-Censits J, Kilpatrick D, Trabulsi EJ, Lallas CD, Chervoneva I, Limentani K, Kennedy B, Kessler S, Gomella L, Antonarakis ES, Carducci MA, Force T, Kelly WK. A pilot phase II Study of digoxin in patients with recurrent prostate cancer as evident by a rising PSA. AMERICAN JOURNAL OF CANCER THERAPY AND PHARMACOLOGY 2014; 2:21-32. [PMID: 25580468 PMCID: PMC4287984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
BACKGROUND Digoxin was found to inhibit prostate cancer (PCa) growth via the inhibition of HIF-1α synthesis in a mouse model. We hypothesized that a therapeutic dose of digoxin could inhibit human PCa growth and disease progression. METHODS An open label, single arm pilot study was performed. Patients (pts) with non-metastatic, biochemically relapsed PCa with prostate specific antigen doubling time (PSADT) of 3-24 months and no hormonal therapy within the past 6 months were enrolled. All pts had testosterone > 50 ng/dL at baseline. Digoxin was taken daily with dose titration to achieve a target therapeutic level (0.8 - 2 ng/ml); patients had routine follow-up including cardiac monitoring with 12-lead electrocardiograms (ECGs) and digoxin levels. The primary endpoint was the proportion of pts at 6 months post-treatment with a PSADT ≥ 200% from the baseline. HIF-1α downstream molecule vascular endothelial growth factor (VEGF) was measured in plasma. RESULTS Sixteen pts were enrolled and 14 pts finished the planned 6 months of treatment. Twenty percent (3/15) of the pts had PSA decrease >25% from baseline with a medium duration of 14 months. At 6 months, 5 of 13 (38%) pts had PSADT ≥ 200% of the baseline PSADT and were continued on study for an additional 24 weeks of treatment. Two patients had durable PSA response for more than 1 year. Digoxin was well tolerated with possible relation of one grade 3 back pain. No patients had evidence of digoxin toxicity. The digoxin dose was lowered in 2 patients for significant ECGs changes (sinus bradycardia and QT prolongation), and there were probable digoxin-related ECG changes in 3 patients. Plasma VEGF was detected in 4 (25%) patients. CONCLUSIONS Digoxin was well tolerated and showed a prolongation of PSDAT in 38% of the patients. However, there was no significant difference comparing that of similar patients on placebo from historical data. Digoxin at the dose used in this study may have limited benefit for patients with biochemically relapsed prostate cancer.
Collapse
Affiliation(s)
- Jianqing Lin
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Tingting Zhan
- Department of Pharmacology and Experimental Therapeutics (Biostatistics), Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Danielle Duffy
- Department of Medicine (Cardiology), Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Jean Hoffman-Censits
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Deborah Kilpatrick
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Edouard J. Trabulsi
- Department of Urology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Costas D. Lallas
- Department of Urology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Inna Chervoneva
- Department of Pharmacology and Experimental Therapeutics (Biostatistics), Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Kimberly Limentani
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Brooke Kennedy
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Sarah Kessler
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Leonard Gomella
- Department of Medicine (Cardiology), Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | | | | | - Thomas Force
- Department of Medicine (Cardiology), Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Wm. Kevin Kelly
- Department of Medical Oncology, Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|