A Contemporary Update on Sipuleucel-T for Men with Metastatic Castrate-Resistant Prostate Cancer

Despite warranted concerns regarding the overdiagnosis and overtreatment of many cases of biologically indolent prostate cancer, prostate cancer remains the second leading cause of cancer-related death in the United States behind only lung cancer.1 With current treatment paradigms, nearly all patients who die of prostate cancer first receive androgen-deprivation therapy and then progress to castrate-resistant prostate cancer.

Written by: Christopher J.D. Wallis, MD, PhD and Zachary Klaassen, MD, MSc
References: 1. Siegel, R. L., & Miller, K. D. (2018). Jemal A (2018) cancer statistics. Ca Cancer J Clin68(1), 7-30.
2. Lowrance, William T., Mohammad Hassan Murad, William K. Oh, David F. Jarrard, Matthew J. Resnick, and Michael S. Cookson. "Castration-resistant prostate cancer: AUA Guideline Amendment 2018." The Journal of urology 200, no. 6 (2018): 1264-1272.
3. Goldman, Bruce, and Laura DeFrancesco. "The cancer vaccine roller coaster." Nature biotechnology 27, no. 2 (2009): 129-139.
4. Kantoff, Philip W., Celestia S. Higano, Neal D. Shore, E. Roy Berger, Eric J. Small, David F. Penson, Charles H. Redfern et al. "Sipuleucel-T immunotherapy for castration-resistant prostate cancer." New England Journal of Medicine 363, no. 5 (2010): 411-422.
5. Higano, Celestia S., Andrew J. Armstrong, A. Oliver Sartor, Nicholas J. Vogelzang, Philip W. Kantoff, David G. McLeod, Christopher M. Pieczonka et al. "Real‐world outcomes of sipuleucel‐T treatment in PROCEED, a prospective registry of men with metastatic castration‐resistant prostate cancer." Cancer 125, no. 23 (2019): 4172-4180.
6. Anassi, Enock, and Uche Anadu Ndefo. "Sipuleucel-T (provenge) injection: the first immunotherapy agent (vaccine) for hormone-refractory prostate cancer." Pharmacy and Therapeutics 36, no. 4 (2011): 197.

Indications: External Urinary Catheters

The use of an external urine collection device (EUCD) is an effective way to manage and collect urine leakage in men and women who have urinary incontinence. However, these devices are not indicated for the management of urinary obstruction or urinary retention. The 2009 CDC guidelines noted that an EUCD is an alternative to an indwelling urinary (Foley) catheter in male patients without urinary retention or bladder outlet obstruction.   

Appropriate use:

  • Male or female patients who experience urinary incontinence (UI) without urinary retention including long term care residents in nursing homes, patients who are obese and have limited movement, and those patients with UI secondary to neurogenic lower urinary tract dysfunction (NLUTD) without sensory awareness due to paralyzing spinal disorders such as spinal cord injury, transverse myelitis, or progressive multiple sclerosis.
  • Patient/caregiver requests for an external device to manage and collect urine leakage.
  • For use in a male patient who has undergone prostate surgery (i.e. post-prostatectomy) who is experiencing stress incontinence who needs a containment system to return to work or usual activities (e.g., golfing).
  • Daily (not hourly) measurement of urine volume that is required (e.g. hospitalized patient) and cannot be assessed by other volume and urine collection strategies in acute care situations (e.g. acute renal failure work-up, bolus diuretics, fluid management in respiratory failure).
  • Single 24-hour or random nonsterile urine sample for diagnostic tests that cannot be obtained by other urine collection strategies.
  • To reduce or minimize acute, severe pain that is a result of movement when other urine management strategies are difficult (e.g. turning patient to remove an absorbent pad causes pain).
  • Managing overactive bladder symptoms and improving comfort in palliative care patients.
  • Use during the night to promote restful sleep and to reduce the risk for falls by minimizing the need to get up to urinate.

Inappropriate use:

  • Any type of urinary retention (acute or chronic, with or without bladder outlet obstruction).
  • Any use in an uncooperative patient expected to frequently manipulate catheters because of such behavior issues as delirium and dementia.
  • Patient or family request in a patient who is continent when there are alternatives for urine containment (e.g. commode, urinal, or bedpan).
  • A need for a sterile urine sample for diagnostic tests where specimen obtained from an EUCD is not sterile.


Written by: Diane K. Newman, DNP, ANP-BC, FAAN, Adjunct Professor of Urology in Surgery, Research Investigator Senior, Perelman School of Medicine, Co-Director, Penn Center for Continence and Pelvic Health, Division of Urology, University of Pennsylvania, Philadelphia, Pennsylvania

April 2020


Written by: Diane K. Newman, DNP, ANP-BC, FAAN
References: 1. Conway, Laurie J., and Elaine L. Larson. "Guidelines to prevent catheter-associated urinary tract infection: 1980 to 2010." Heart & lung 41, no. 3 (2012): 271-283.
2. Deng, Donna Y. "Urologic Devices." In Clinical Application of Urologic Catheters, Devices and Products, pp. 173-220. Springer, Cham, 2018.
3. Geng, V., H. Cobussen-Boekhorst, H. Lurvink, I. Pearce, and S. Vahr. "Evidence-based guidelines for best practice in urological health care: male external catheters in adults urinary catheter management." Arnhem: European Association of Urology Nurses (2016).
4. Gould, Carolyn V., Craig A. Umscheid, Rajender K. Agarwal, Gretchen Kuntz, David A. Pegues, and Healthcare Infection Control Practices Advisory Committee. "Guideline for prevention of catheter-associated urinary tract infections 2009." Infection Control & Hospital Epidemiology 31, no. 4 (2010): 319-326.
5. Gray, Mikel, Claudia Skinner, and Wendy Kaler. "External collection devices as an alternative to the indwelling urinary catheter: evidence-based review and expert clinical panel deliberations." Journal of Wound, Ostomy, and Continence Nursing 43, no. 3 (2016): 301.
6. Hooton, Thomas M., Suzanne F. Bradley, Diana D. Cardenas, Richard Colgan, Suzanne E. Geerlings, James C. Rice, Sanjay Saint et al. "Diagnosis, prevention, and treatment of catheter-associated urinary tract infection in adults: 2009 International Clinical Practice Guidelines from the Infectious Diseases Society of America." Clinical infectious diseases 50, no. 5 (2010): 625-663.
7. Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
8. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.
9. Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
10. Tenke, Peter, Bela Kovacs, Truls E. Bjerklund Johansen, Tetsuro Matsumoto, Paul A. Tambyah, and Kurt G. Naber. "European and Asian guidelines on management and prevention of catheter-associated urinary tract infections." International journal of antimicrobial agents 31 (2008): 68-78.

The Risks of Delaying Kidney Cancer Treatment During COVID-19

The rapid spread of Coronavirus Disease 2019 (COVID-19), caused by the betacoronavirus SARS-CoV-2, has had dramatic effects throughout the world on healthcare systems with impacts far beyond the patients actually infected with COVID-19.

Patients who manifest severe forms of COVID-19 requiring respiratory support typically require this for prolonged durations, with a mean of 13 days of respiratory support reported by the China Medical Treatment Expert Group for COVID-19.1 This lengthy requirement for ventilator support and ICU resources, exacerbated by relatively little excess health system capacity to accommodate epidemics, means that healthcare systems can (and have in the case of many hospitals in Italy) become overwhelmed relatively quickly. In an effort to conserve hospital resources, on March 13th the American College of Surgeons recommended that health systems, hospitals, and surgeons should attempt to minimize, postpone, or outright cancel electively scheduled operations.2 This was done with the primary goal to immediately decrease the use of items essential for the care of patients with COVID-19 including ICU beds, ventilators, personal protective equipment, and terminal cleaning supplies. On March 17th, the American College of Surgeon then provided further guidance on the triage of non-emergent surgeries, including an aggregate assessment of the risk incurred from surgical delays of six to eight weeks or more as compared to the risk (both to the patient and the healthcare system) of proceeding with the operation.3 In the UK, all non-urgent elective surgical procedures have been put on hold for three months to use all of those clinical resources to care for patients with COVID-19.

Most bodies, including the American College of Surgeons, have recommended proceeding with most cancer surgeries. Thus, clinicians and patients must carefully weigh the benefit of proceeding with cancer treatment as scheduled, the risks of COVID-19 to the individual patient, to healthcare workers caring for patients potentially infected with COVID-19, and the need to conserve health care resources. A severe SARS-CoV-2 phenotype is seen more commonly in men and older, more comorbid patients.4 Baseline characteristics among 1,591 patients admitted to the ICU in the Lombardy Region, Italy, showed that the median age was 63 years (IQR 56-70), 82% were male, 68% had ≥1 comorbidity, 88% required ventilator support, and the mortality rate was 26%, with a large proportion requiring ongoing ICU level care at the time of data cut-off.5 Work from China demonstrated that patients with cancer had a higher incidence of COVID-19 infection than expected in the general population and had a more severe manifestation of the disease with a significantly higher proportion requiring invasive ventilation in the ICU or death.6 Patients at increased risk of COVID-19 are comparable to the kidney cancer population, namely more likely male, hypertensive, and with more than one comorbidity.5


Thus, considering the differences in the natural history of different cancers may meaningfully change this balance of risks and benefits. Kidney cancer has a wide range of phenotypic presentations ranging from very indolent, incidentally diagnosed small renal masses to large, symptomatic tumors with vascular or adjacent organ invasion and de novo metastatic disease. Previous articles in the UroToday Kidney Cancer Today Center of Excellence have discussed the Epidemiology and Etiology of Kidney Cancer and particular nuances of staging and management for Malignant Renal Tumors. The vast majority of patients have localized disease at the time of presentation. According to Siegel et al., 65% of all patients diagnosed with kidney and renal pelvis tumors between 2007 and 2013 had localized disease at the time of presentation while 16% had regional spread, and 16% had evidence of distant, metastatic disease.7

While the effect of delays in surgical intervention has been most thoroughly explored in muscle-invasive bladder cancer and prostate cancer in the urologic literature, both data and inference may help guide the management of patients with kidney cancer at this time. A single previous narrative review has assessed this question,8 however, conclusions from this study are limited. In the absence of data, the guidance of care relies on expert opinion, including a collaborative review pre-published in European Urology (Wallis et al.). This article will discuss the impact of potential delays among patients with kidney cancer, providing recommendations as to who can safely defer treatment until after the pandemic is over versus those that should be treated without delay.

While there are no randomized data, numerous institutional studies have demonstrated both the safety and feasibility of active surveillance (AS) with delayed intervention in patients with small renal masses (SRMs).9,10

Among 457 patients treated at Fox Chase Cancer Center, McIntosh et al. found that rates of delayed intervention were 9%, 22%, 29%, 35%, and 42% at one-, two-, three-, four-, and five-years following diagnosis.11 Importantly, delayed intervention was not associated with overall survival (OS) (hazard ratio [HR] 1.34, 95% confidence interval [CI] 0.79-2.29), and of the 99 patients on AS without delayed intervention for more than five years, only one patient metastasized. Interestingly, they found a median initial linear growth rate was 1.9 mm/year (IQR 0-7) which was not associated with OS.

Data from the University of Michigan has also assessed the effect of delayed resection (at least six months from presentation) after initial surveillance for patients with SRMs.12 In this study, there were 401 patients that underwent early resection and 94 patients (19%) that underwent delayed resection. The median time to resection was 84 days (IQR 59-121) in the early intervention group and 386 days (IQR 272-702) in the delayed intervention group. Importantly, there was no difference in adverse final pathology (grade 3-4, papillary type 2, sarcomatoid histology, angiomyolipoma with epithelioid features, or stage ≥ pT3) comparing those that underwent early vs late intervention.

Among 497 patients in the DISSRM registry, 223 (43%) chose AS and 274 (57%) chose primary intervention with 21 (9%) eventually undergoing delayed intervention after a period of AS. There was no difference in cancer-specific survival (CSS) at five-years between the groups (primary intervention: 99%; AS 100%, p=0.3) though patients choosing surveillance had lower five-year OS (75% vs 92%), likely attributable to comorbidity which drove their initial selection of surveillance.

Taken together, there is robust data supporting AS for masses < 4cm, even up to five years after the initial diagnosis, without age restriction.13,14 Thus, delays in the diagnosis and management of these patients during the COVID-19 pandemic is unlikely to meaningfully affect their outcomes.

Although the data is less robust, several studies have assessed the impact of a surgical delay for larger localized kidney tumors (≥pT1b). An institutional cohort from Memorial Sloan Kettering Cancer Center examined 1,278 patients who underwent radical or partial nephrectomy with renal masses > 4 cm between 1995 and 2013. The authors tested the association between surgical wait time and disease upstaging at the time of surgery, as well as two- and five-year recurrence rates.15 Among these patients, 267 (21%) had a surgical wait time of more than three months, including 82 patients (6%) with a wait time of more than six months. On multivariable analysis, the surgical wait time was not associated with disease upstaging, recurrence or CSS, but longer wait time was associated with worse OS (HR 1.17, 95% CI 1.08-1.27), potentially reflecting comorbidity which necessitated the initial delays.

Similarly, an institutional cohort from the Fox Chase Cancer Center of patients with cT1b/cT2 renal masses assessed the safety of active surveillance.16 Twenty-three patients (34%) underwent delayed intervention. Over a median follow-up of 32 months (range 6-119), no patients progressed to metastatic disease or died of kidney cancer. The median linear growth rate was 0.34 cm/year (range 0-1.48) suggesting that delays of months to years are unlikely to affect the resectability of these tumors.

Finally, a retrospective review of 722 patients undergoing partial or radical nephrectomy for relatively large kidney tumors (mean tumor size of 6.4 +/- 4.4cm; 64.7% ≥pT1b and 49.0% ≥pT2) from Stec et al. assessed the effect of delays to surgery on survival.17 They found that the mean time from initial visit to surgery was 1.2 months (range 0-30 months) and that 64.1% of patients underwent surgery within 30 days of initial visit and 94.3% within three months. The authors found no difference in OS for patients receiving early vs. late surgery, whether using a threshold of one month (p=0.87), two months (p=0.46), three months (p=0.71), or six months (p=0.75). However, T-stage was a significant predictor of recurrence-free survival (RFS), independent of time to surgery.

There are essentially no available data on the effect of delays in treating patients with locally advanced kidney cancer. However, several large institutional studies have described timing of preoperative imaging for assessing renal vein/inferior vena cava (IVC) thrombus, which may guide the urgency of surgical timing. While Woodruff et al. recommended the longest interval between imaging (CT/MRI) and surgery being no longer than 30 days,18 studies from the Mayo Clinic and Berlin, Germany report a median interval from imaging to resection of 4 and 16 days, respectively.19,20 As a result, the collaborative review pre-published in European Urology suggested that these patients should be prioritized for intervention given the locally advanced nature of their disease, unknown risk of delayed resection, and potential for significant symptomatic complications including bleeding and IVC occlusion.

In contrast, in patients with known metastatic kidney cancer, cytoreductive nephrectomy during the ongoing pandemic was not recommended in this collaborative review on account of the evidence from the CARMENA trial which failed to demonstrate a survival benefit to the addition of cytoreductive to systemic therapy with sunitinib.21 Additionally, the SURTIME trial found that upfront systemic therapy followed by cytoreductive nephrectomy was associated with longer survival than patients who received upfront cytoreduction followed by systemic therapy.22

The question of when to initiate systemic therapy in patients with metastatic kidney cancer is also pertinent during this pandemic and depends on symptoms, patient comorbidities, and tumor risk stratification. In treatment-naïve patients with favorable and intermediate-risk disease who are asymptomatic or minimally symptomatic with limited disease burden, a number of studies, including two narrative reviews23,24 and two prospective trials,25,26 have assessed the safety and feasibility of delaying the initiation of systemic therapy. In the largest prospective study of surveillance in patients with metastatic kidney cancer, Rini et al. enrolled fifty-two asymptomatic patients in a Phase II trial.26 All but one patient had favorable (23%) or intermediate (75%) risk disease according to IMDC criteria with the vast majority (80%) of patients having only one or two organ sites with metastases. The median time on surveillance was 14.9 months and 37 of 43 patients experiencing RECIST-defined disease progression started systemic treatment. The median PFS and OS from the start of surveillance was 9.4 and 44.5 months, respectively. In a smaller study of 15 patients who received CN followed by observation until progression, Wong et al. found a median time to progression of eight weeks and a median OS of 25 months.25 Neither of these studies have compared outcomes with patients who started systemic therapy upfront. However, this has been assessed in two large retrospective studies. Among a European cohort of patients ineligible for active surveillance, Iacovelli and colleagues found that a delay of more than six weeks in the initiation of systemic therapy did not significantly affect the cancer-specific outcomes.27 Further, delayed treatment initiation was not independently associated with worse OS in an analysis based on the National Cancer Database utilizing multivariable logistic regression analyses.28

There is no evidence to guide the choice of systemic therapy during the COVID-19 pandemic and thus, standard guideline recommendations should prevail. However, at least theoretically, the risk of severe adverse reactions associated with immunotherapy using checkpoint inhibitors, as well as the requirement for infusion, may preferentially support the use of VEGF-targeted therapy with the convenience of oral administration.

Written by: Zachary Klaassen, MD, MSc, Assistant Professor of Urology, Georgia Cancer Center, Augusta University/Medical College of Georgia, Atlanta, Georgia

Published Date: April 2020

Written by: Zachary Klaassen, MD, MSc
References: 1. Guan, Wei-jie, Zheng-yi Ni, Yu Hu, Wen-hua Liang, Chun-quan Ou, Jian-xing He, Lei Liu et al. "Clinical characteristics of coronavirus disease 2019 in China." New England Journal of Medicine (2020).
2. March 13, Online, and 2020. “COVID-19: Recommendations for Management of Elective Surgical Procedures.” American College of Surgeons. Accessed April 20, 2020. https://www.facs.org/covid-19/clinical-guidance/elective-surgery.
3. March 17, Online, and 2020. “COVID-19: Guidance for Triage of Non-Emergent Surgical Procedures.” American College of Surgeons. Accessed April 20, 2020. https://www.facs.org/covid-19/clinical-guidance/triage.
4.COVID, CDC, and Response Team. "Severe outcomes among patients with coronavirus disease 2019 (COVID-19)—United States, February 12–March 16, 2020." MMWR Morb Mortal Wkly Rep 69, no. 12 (2020): 343-346.
5. Grasselli, Giacomo, Alberto Zangrillo, Alberto Zanella, Massimo Antonelli, Luca Cabrini, Antonio Castelli, Danilo Cereda et al. "Baseline characteristics and outcomes of 1591 patients infected with SARS-CoV-2 admitted to ICUs of the Lombardy region, Italy." JAMA (2020).
6. Liang, Wenhua, Weijie Guan, Ruchong Chen, Wei Wang, Jianfu Li, Ke Xu, Caichen Li et al. "Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China." The Lancet Oncology 21, no. 3 (2020): 335-337.
7. Siegel, Rebecca L., and Kimberly D. Miller. "Jemal A (2018) cancer statistics." Ca Cancer J Clin 68, no. 1 (2018): 7-30.
8. Bourgade, Vincent, Sarah J. Drouin, David R. Yates, Jerôme Parra, Marc-Olivier Bitker, Olivier Cussenot, and Morgan Rouprêt. "Impact of the length of time between diagnosis and surgical removal of urologic neoplasms on survival." World journal of urology 32, no. 2 (2014): 475-479.
9. Mir, Maria Carmen, Umberto Capitanio, Riccardo Bertolo, Idir Ouzaid, Maciej Salagierski, Maximilian Kriegmair, Alessandro Volpe, Michael AS Jewett, Alexander Kutikov, and Phillip M. Pierorazio. "Role of active surveillance for localized small renal masses." European urology oncology 1, no. 3 (2018): 177-187.
10. Sanchez, Alejandro, Adam S. Feldman, and A. Ari Hakimi. "Current management of small renal masses, including patient selection, renal tumor biopsy, active surveillance, and thermal ablation." Journal of Clinical Oncology 36, no. 36 (2018): 3591.
11. McIntosh, Andrew G., Benjamin T. Ristau, Karen Ruth, Rachel Jennings, Eric Ross, Marc C. Smaldone, David YT Chen et al. "Active surveillance for localized renal masses: tumor growth, delayed intervention rates, and> 5-yr clinical outcomes." European urology 74, no. 2 (2018): 157-164.
12. Hawken, Scott R., Naveen K. Krishnan, Sapan N. Ambani, Jeffrey S. Montgomery, Elaine M. Caoili, James H. Ellis, Lakshmi P. Kunju et al. "Effect of delayed resection after initial surveillance and tumor growth rate on final surgical pathology in patients with small renal masses (SRMs)." In Urologic Oncology: Seminars and Original Investigations, vol. 34, no. 11, pp. 486-e9. Elsevier, 2016.
13. Kutikov, Alexander. "Surveillance of Small Renal Masses in Young Patients: A Viable Option in the Appropriate Candidate." European urology focus 2, no. 6 (2016): 567-568.
14. Pierorazio, Phillip M., Michael H. Johnson, Mark W. Ball, Michael A. Gorin, Bruce J. Trock, Peter Chang, Andrew A. Wagner, James M. McKiernan, and Mohamad E. Allaf. "Five-year analysis of a multi-institutional prospective clinical trial of delayed intervention and surveillance for small renal masses: the DISSRM registry." European urology 68, no. 3 (2015): 408-415.
15. Mano, Roy, Emily A. Vertosick, Abraham Ari Hakimi, Itay A. Sternberg, Daniel D. Sjoberg, Melanie Bernstein, Guido Dalbagni, Jonathan A. Coleman, and Paul Russo. "The effect of delaying nephrectomy on oncologic outcomes in patients with renal tumors greater than 4 cm." In Urologic Oncology: Seminars and Original Investigations, vol. 34, no. 5, pp. 239-e1. Elsevier, 2016.
16. Mehrazin, Reza, Marc C. Smaldone, Alexander Kutikov, Tianyu Li, Jeffrey J. Tomaszewski, Daniel J. Canter, Rosalia Viterbo, Richard E. Greenberg, David YT Chen, and Robert G. Uzzo. "Growth kinetics and short-term outcomes of cT1b and cT2 renal masses under active surveillance." The Journal of urology 192, no. 3 (2014): 659-664.
17. Stec, Andrew A., Benjamin J. Coons, Sam S. Chang, Michael S. Cookson, S. Duke Herrell, Joseph A. Smith Jr, and Peter E. Clark. "Waiting time from initial urological consultation to nephrectomy for renal cell carcinoma—does it affect survival?." The Journal of urology 179, no. 6 (2008): 2152-2157.
18. Woodruff, Daniel Y., Peter Van Veldhuizen, Gregory Muehlebach, Phillip Johnson, Timothy Williamson, and Jeffrey M. Holzbeierlein. "The perioperative management of an inferior vena caval tumor thrombus in patients with renal cell carcinoma." In Urologic Oncology: Seminars and Original Investigations, vol. 31, no. 5, pp. 517-521. Elsevier, 2013.
19. Psutka, Sarah P., Stephen A. Boorjian, Robert H. Thompson, Grant D. Schmit, John J. Schmitz, Thomas C. Bower, Suzanne B. Stewart, Christine M. Lohse, John C. Cheville, and Bradley C. Leibovich. "Clinical and radiographic predictors of the need for inferior vena cava resection during nephrectomy for patients with renal cell carcinoma and caval tumour thrombus." BJU international 116, no. 3 (2015): 388-396.
20. Adams, Lisa C., Bernhard Ralla, Yi-Na Y. Bender, Keno Bressem, Bernd Hamm, Jonas Busch, Florian Fuller, and Marcus R. Makowski. "Renal cell carcinoma with venous extension: prediction of inferior vena cava wall invasion by MRI." Cancer Imaging 18, no. 1 (2018): 17.
19. Méjean, Arnaud, Alain Ravaud, Simon Thezenas, Sandra Colas, Jean-Baptiste Beauval, Karim Bensalah, Lionnel Geoffrois et al. "Sunitinib alone or after nephrectomy in metastatic renal-cell carcinoma." New England Journal of Medicine 379, no. 5 (2018): 417-427.
21. Bex, Axel, Peter Mulders, Michael Jewett, John Wagstaff, Johannes V. Van Thienen, Christian U. Blank, Roland Van Velthoven et al. "Comparison of immediate vs deferred cytoreductive nephrectomy in patients with synchronous metastatic renal cell carcinoma receiving sunitinib: the SURTIME randomized clinical trial." JAMA oncology 5, no. 2 (2019): 164-170.
23. Pickering, Lisa M., Mohammed O. Mahgoub, and Deborah Mukherji. "Is observation a valid strategy in metastatic renal cell carcinoma?." Current opinion in urology 25, no. 5 (2015): 390-394.
24. Nizam, Amanda, Jonah A. Schindelheim, and Moshe C. Ornstein. "The role of active surveillance and cytoreductive nephrectomy in metastatic renal cell carcinoma." Cancer Treatment and Research Communications (2020): 100169.
25. Wong, Alvin S., Kian-Tai Chong, Chin-Tiong Heng, David T. Consigliere, Kesavan Esuvaranathan, Khai-Lee Toh, Benjamin Chuah, Robert Lim, and James Tan. "Debulking nephrectomy followed by a “watch and wait” approach in metastatic renal cell carcinoma." In Urologic Oncology: Seminars and Original Investigations, vol. 27, no. 2, pp. 149-154. Elsevier, 2009.
26. Rini, Brian I., Tanya B. Dorff, Paul Elson, Cristina Suarez Rodriguez, Dale Shepard, Laura Wood, Jordi Humbert et al. "Active surveillance in metastatic renal-cell carcinoma: a prospective, phase 2 trial." The Lancet Oncology 17, no. 9 (2016): 1317-1324.
27. Iacovelli, Roberto, Luca Galli, Ugo De Giorgi, Camillo Porta, Franco Nolè, Paolo Zucali, Roberto Sabbatini et al. "The effect of a treatment delay on outcome in metastatic renal cell carcinoma." In Urologic Oncology: Seminars and Original Investigations, vol. 37, no. 8, pp. 529-e1. Elsevier, 2019.
28. Woldu, Solomon L., Justin T. Matulay, Timothy N. Clinton, Nirmish Singla, Yuval Freifeld, Oner Sanli, Laura-Maria Krabbe et al. "Incidence and outcomes of delayed targeted therapy after cytoreductive nephrectomy for metastatic renal-cell carcinoma: a nationwide cancer registry study." Clinical genitourinary cancer 16, no. 6 (2018): e1221-e1235.

After POUT: Implications of Perioperative Chemotherapy for Upper Tract Urothelial Carcinoma

Before the POUT Trial

Before the POUT trial, the largest randomized Phase III trial assessing perioperative chemotherapy for locally advanced UTUC, decisions in management relied primarily on retrospective studies and expert opinion. Furthermore, the data from these studies are conflicting regarding the efficacy of perioperative chemotherapy. In 2009, Hellenthal and colleagues5 used an international collaborative database to assess the utilization and outcomes of adjuvant chemotherapy after radical nephroureterectomy in patients with UTUC. They identified 1,390 patients who underwent nephroureterectomy for nonmetastatic UTUC between 1992 and 2006. Of these cases, 39% were classified as high-risk (pT3N0, pT4N0 and/or lymph node-positive). For the analysis, these patients were divided into two groups, including those who did and did not receive adjuvant chemotherapy, and stratified by gender, age group, performance status, and tumor grade and stage. Among the 542 high-risk patients, 22% received adjuvant chemotherapy, which was more commonly administered in the context of increased tumor grade and stage (p <0.001). The median survival for the entire cohort was 24 months (range 0 to 231 months), and there was no significant difference in all-cause death (chemotherapy vs none: hazard ratio [HR] 1.06, 95% confidence interval [CI] 0.80-1.40) or cancer-specific death (chemotherapy vs none: HR 1.26, 95% CI 0.93-1.71) between patients who did and did not receive adjuvant chemotherapy.

A subsequent study from the European Association of Urology – Young Academic Urologists and the Upper Tract Urothelial Collaboration also assessed adjuvant chemotherapy after radical nephroureterectomy.6 This study collated data from 15 centers (1,544 patients) between 2000 and 2015; patients were required to have pT2-4N0/Nx or N+ disease and had undergone a radical nephroureterectomy. The primary analysis used 1:1 propensity score matching, with inverse probability of treatment weighting with a primary endpoint of overall survival (OS). Among the 1,544 patients included, 312 received adjuvant chemotherapy and 1,232 underwent observation. In the matched analysis, there was no difference observed in OS between adjuvant chemotherapy and observation (HR 1.14, 95% CI 0.91-1.43; P = 0.268). Furthermore, a six‐month landmark analysis demonstrated little impact of early events on the treatment effect on OS, with an HR of 1.28 (95% CI 1.00–1.64; p = 0.051).

In contrast to these two studies that failed to demonstrate a benefit to adjuvant chemotherapy, a systematic review and meta-analysis published in 2014 assessed the role of adjuvant and neoadjuvant chemotherapy for patients with UTUC.7 Not surprisingly, this systematic review found no randomized trials investigated the role of adjuvant chemotherapy for UTUC. There was one prospective study comprising 36 patients that investigated adjuvant carboplatin-paclitaxel. Furthermore, there were nine retrospective studies (totaling 482 patients) among patients receiving cisplatin-based or non-cisplatin-based adjuvant chemotherapy after nephroureterectomy compared to 1,300 patients receiving nephroureterectomy alone. Across the three cisplatin-based studies, the pooled hazard ratio for OS was 0.43 (95% CI, 0.21-0.89; p=0.023) favoring adjuvant cisplatin compared with those who received surgery alone. For disease-free survival (DFS), the pooled HR across two studies was 0.49 (95% CI, 0.24-0.99; p=0.048) favoring adjuvant cisplatin. Among the non-cisplatin-based chemotherapy studies, there was no benefit for neoadjuvant chemotherapy. For the neoadjuvant chemotherapy section of the systematic review, the authors identified two Phase II trials that demonstrated favorable pathologic downstaging rates (60%-75% downstaging to ≤pT1N0). Across two retrospective studies investigating neoadjuvant chemotherapy, there was a disease-specific survival (DSS) benefit for neoadjuvant chemotherapy, with a pooled HR of 0.41 (95% CI, 0.22-0.76; p=0.005).


The POUT Trial

The POUT trial was a Phase III, parallel group, open-label, randomized controlled trial done at 71 National Health Service (NHS) hospitals in the United Kingdom.8 Eligible patients were ≥16 years, had received a radical nephroureterectomy for UTUC, were postoperatively staged with either muscle-invasive (pT2–pT4, pNany) or lymph node-positive (pTany, pN1–3) M0 disease with predominantly transitional cell carcinoma histology, and were fit to receive adjuvant chemotherapy within 90 days of surgery. Patients also had to have a glomerular filtration rate (GFR) of ≥30 mL/min. Prespecified stratification factors included platinum chemotherapy agent (cisplatin vs carboplatin), preoperative radiologically or pathologically assessed nodal involvement (N0 vs N1 vs N2 vs N3), the status of surgical margins (positive vs negative), and treatment center. Patients were randomized 1:1 to receive either surveillance or adjuvant chemotherapy: four 21-day cycles of platinum-based chemotherapy (cisplatin 70 mg/m2) within 14 days of randomization; gemcitabine (1000 mg/m2) given on days one and eight of each cycle. Patients with impaired renal function (GFR ≥30 mL/min and <50 mL/min) received carboplatin rather than cisplatin.

Patients were followed at 3, 6, 9, and 12 months, then every 6 months to 36 months from randomization, and annually thereafter. The radiographic assessment included a CT of the thorax, abdomen, and pelvis at 3, 6, 9, 12, 18, 24, 30, and 36 months, then annually to 60 months. Cystoscopy was done every 6 months to 24 months, then annually up to 60 months after surgery. Toxicity was assessed by CTCAE v4.  Furthermore, this was the first trial in UTUC to collect patient-reported outcomes: patients filled out the EORTC quality-of-life of cancer patients questionnaire (QLQ-C30) and the EuroQol five dimensions five levels questionnaire (EQ-5D-5L) at baseline and before cycle three and at 3, 6, 12, and 24 months after randomization. The primary endpoint of this trial was DFS defined as time from randomization to either first recurrence in the tumor bed, first metastasis, or death from any cause. Secondary endpoints included metastasis-free survival (MFS), OS, treatment compliance, acute toxicity, late toxicity, and patient-reported quality of life. The trial was powered to detect a hazard ratio of 0.65 (i.e. improvement in three-year DFS from 40% to 55%; 2-sided alpha = 5%, 80% power) with Peto-Haybittle (p<0.001) early stopping rules for efficacy and inefficacy.

There were 261 patients included in the trial between June 19, 2012 and November 8, 2017 at 57 of the 71 participating centers in the UK, including 129 patients randomized to surveillance and 132 to chemotherapy; 260 patients were included in the intention to treat analysis. In October 2017, the independent trial oversight committees recommended POUT close to recruitment as data collected to date met the early stopping rule for efficacy. Included patients were a median 68.5 years of age (IQR 62.0-74.1 years). With respect to tumor characteristics, 94% of patients had pT2-T3 disease and 91% were N0. The median follow-up was 30.3 months (IQR 18.0-47.5 months). There were 7 of 131 patients allocated to chemotherapy that did not start treatment and 75% of those that started chemotherapy received four cycles. There were 60 (47%) DFS events in the surveillance cohort and 35 (27%) in the chemotherapy cohort; as such, the unadjusted HR was 0.45 (95% CI 0.30-0.68) in favor of chemotherapy (log-rank p = 0.0001). The three-year DFS rate was 46% for surveillance (95% CI 36-56) and 71% for chemotherapy (95% CI 61-78). MFS also favored chemotherapy, with an HR of 0.48 (95% CI 0.31-0.74, log-rank p = 0.0007), and the three-year event-free rates were 53% (95% CI 42-63) for those on surveillance and 71% (95% CI 60-79) for those receiving chemotherapy. Currently, there have been 62 deaths recorded (38 for surveillance, 24 for chemotherapy) and the OS analysis is planned for when 88 deaths occur, or all patients have ≥2 years of follow-up.

Grade ≥3 toxicities were reported in 44% of chemotherapy patients and 4% surveillance patients (p < 0.0001). During the treatment period, the most common grade ≥3 toxicities in chemotherapy patients were neutropenia (36%) and thrombocytopenia (10%). Analysis of late toxicity is planned after a two-year follow-up of all patients. Quality of life questionnaires were returned by 95% of patients at baseline (95% of those allocated to surveillance and 95% of those allocated to chemotherapy), 82% at three months (81% of those allocated to surveillance and 82% of those allocated to chemotherapy), and 70% at 12 months (70% of those allocated to surveillance and 70% of those allocated to chemotherapy). The mean overall global health status score at baseline was 77% (standard deviation 19) for the chemotherapy group and 76% (standard deviation 19) for the surveillance group. Subsequently, the overall global health status was lower during chemotherapy (before cycle 3) and immediately afterward (at three months) in participants allocated chemotherapy versus surveillance, however the difference resolved by six months of follow-up.

Implications and Future Considerations Following the POUT Trial

Given the rarity of UTUC and the clinical equipoise for conducting a perioperative chemotherapy trial in this disease state, the trialist and the patients involved in this study are to be congratulated for completing this important Phase III trial. As has been discussed amongst thought leaders in the field and summarized in a discussion between Dr. Ashish Kamat and trial lead Dr. Alison Birtle, this is practice-changing data: all patients with locally advanced UTUC after radical nephrectomy should be considered for receipt of adjuvant cisplatin-based chemotherapy.

There are several important considerations as we move into the post-POUT trial era of treating locally advanced UTUC. First, patients with adequate renal function should receive the combination of cisplatin-gemcitabine adjuvant chemotherapy. Typically, a GFR of <60 mL/min is deemed “cisplatin-ineligible”, however, the POUT trial included patients with GFRs as low as 50 mL/min, which is standard practice in the UK. However, even for those with a GFR < 50 mL/min that received the combination of carboplatin-gemcitabine (n=96), there was still a non-significant DFS benefit (HR 0.66, 95% CI 0.35-1.26).8 As such, essentially all pT2–pT4, Nany or pTany, N1–3M0 patients should be considered for an adjuvant chemotherapy regimen that fits their performance status and post-radical nephroureterectomy renal function profile.

Second, the argument for adjuvant chemotherapy as standard of care should only grow stronger as the data matures. Indeed, the benefit of adjuvant chemotherapy was based on DFS and MFS benefit, however with such strong hazard ratios favoring chemotherapy vs surveillance (DFS HR 0.45; MFS HR 0.48) there likely will be a survival benefit with longer follow-up. Furthermore, early survival data reported in the POUT publication favor chemotherapy vs surveillance (38 deaths in surveillance arm; 24 deaths in chemotherapy arm). As such, there is a signal for a more than likely survival benefit based on DFS/MFS benefit and early survival data.

Third, as POUT has demonstrated a survival benefit to perioperative chemotherapy in the adjuvant setting it has re-raised questions regarding the role of neoadjuvant therapy. As nephroureterectomy removes a significant nephron mass, neoadjuvant administration would allow more patients to receive cisplatin, which is known to be more efficacious than carboplatin. However, this must be balanced against issues of poor pre-operative staging in UTUC. Unlike bladder cancer in which TURBT fairly reliably can distinguish superficial from muscle-invasive disease, this distinction is much more difficult in UTUC. Certainly, in patients with obvious invasive disease based on axial imaging and marginal renal function, extrapolation from POUT would suggest that neoadjuvant chemotherapy may be a rational approach.

Finally, the question remains as to how immunotherapy fits into the picture given the POUT data. There are currently no Phase III trials underway to address the role of immunotherapy in the adjuvant treatment of UTUC alone, however, ongoing trials assessing pembrolizumab (Neoadjuvant Pembrolizumab in Combination with Gemcitabine Therapy in Cis-eligible/Ineligible Urothelial Carcinoma Patients) in the perioperative setting for bladder urothelial carcinoma have preplanned subgroup analyses for patients with UTUC. Atezolizumab has shown real-world efficacy in metastatic UTUC, as evaluated in the SAUL study (22% included had UTUC)9, however, what role immunotherapy plays in the perioperative setting for UTUC remains to fully elucidated.

Written by: Zachary Klaassen, MD, MSc
References: 1. Rouprêt, Morgan, Marko Babjuk, Eva Compérat, Richard Zigeuner, Richard J. Sylvester, Maximilian Burger, Nigel C. Cowan et al. "European association of urology guidelines on upper urinary tract urothelial carcinoma: 2017 update." European urology 73, no. 1 (2018): 111-122.
2. Sternberg, Cora N., Iwona Skoneczna, J. Martijn Kerst, Peter Albers, Sophie D. Fossa, Mads Agerbaek, Herlinde Dumez et al. "Immediate versus deferred chemotherapy after radical cystectomy in patients with pT3–pT4 or N+ M0 urothelial carcinoma of the bladder (EORTC 30994): an intergroup, open-label, randomised phase 3 trial." The lancet oncology 16, no. 1 (2015): 76-86.
3. of Trialists, International Collaboration. "International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: long-term results of the BA06 30894 trial." Journal of Clinical Oncology 29, no. 16 (2011): 2171.
4. Vale, C. A. "Advanced Bladder Cancer (ABC). Meta-analysis Collaboration. Adjuvant chemotherapy in invasive bladder cancer: a systematic review and meta-analysis of individual patient data." Eur Urol 48, no. 2 (2005): 189-199.
5. Hellenthal, Nicholas J., Shahrokh F. Shariat, Vitaly Margulis, Pierre I. Karakiewicz, Marco Roscigno, Christian Bolenz, Mesut Remzi et al. "Adjuvant chemotherapy for high risk upper tract urothelial carcinoma: results from the Upper Tract Urothelial Carcinoma Collaboration." The Journal of urology 182, no. 3 (2009): 900-906.
6. Necchi, Andrea, Salvatore Lo Vullo, Luigi Mariani, Marco Moschini, Kees Hendricksen, Michael Rink, Roman Sosnowski et al. "Adjuvant chemotherapy after radical nephroureterectomy does not improve survival in patients with upper tract urothelial carcinoma: a joint study by the European Association of Urology–Young Academic Urologists and the Upper Tract Urothelial Carcinoma Collaboration." BJU international 121, no. 2 (2018): 252-259.
7. Leow, Jeffrey J., William Martin-Doyle, Andre P. Fay, Toni K. Choueiri, Steven L. Chang, and Joaquim Bellmunt. "A systematic review and meta-analysis of adjuvant and neoadjuvant chemotherapy for upper tract urothelial carcinoma." European urology 66, no. 3 (2014): 529-541.
8. Birtle, Alison, Mark Johnson, John Chester, Robert Jones, David Dolling, Richard T. Bryan, Christopher Harris et al. "Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): a phase 3, open-label, randomised controlled trial." The Lancet (2020).
9. Sternberg, Cora N., Yohann Loriot, Nicholas James, Ernest Choy, Daniel Castellano, Fernando Lopez-Rios, Giuseppe L. Banna et al. "Primary results from SAUL, a multinational single-arm safety study of atezolizumab therapy for locally advanced or metastatic urothelial or nonurothelial carcinoma of the urinary tract." European urology 76, no. 1 (2019): 73-81.

Prostate Cancer Early Detection During the COVID-19 Pandemic

Currently, there is a global pandemic surrounding the spread of betacoronavirus SARS-CoV-2 leading to Coronavirus Disease 2019 (COVID-19). The rapid spread to all corners of the globe has had tremendous health and economic implications, including the appropriate allocation of healthcare resources. Considering that hospitals may be overwhelmed quickly given the need for a proportion of patients that require hospitalization with possible ventilator support, there is a necessity to decrease the use of items essential for the care of patients with COVID-19 including ICU beds, ventilators, personal protective equipment, and terminal cleaning supplies. This includes reassessing the priority and implications of treatments, including prostate cancer screening. 

Written by: Zachary Klaassen, MD, MSc
References: 1. Wilson, James Maxwell Glover, Gunnar Jungner, and World Health Organization. "Principles and practice of screening for disease." (1968).

2. Sanda, Martin G., Jeffrey A. Cadeddu, Erin Kirkby, Ronald C. Chen, Tony Crispino, Joann Fontanarosa, Stephen J. Freedland et al. "Clinically localized prostate cancer: AUA/ASTRO/SUO guideline. Part I: risk stratification, shared decision making, and care options." The Journal of urology 199, no. 3 (2018): 683-690.

3. Network NCC. NCCN Clinical Practice Guidelines in Oncology: Prostate Cancer - Version 2.2019. In:2019.

4. Reading, Stephanie R., Kimberly R. Porter, Jin-Wen Y. Hsu, Lauren P. Wallner, Ronald K. Loo, and Steven J. Jacobsen. "Racial and Ethnic Variation in Time to Prostate Biopsy After an Elevated Screening Level of Serum Prostate-specific Antigen." Urology 96 (2016): 121-127.

5. Grummet, Jeremy P., Mahesha Weerakoon, Sean Huang, Nathan Lawrentschuk, Mark Frydenberg, Daniel A. Moon, Mary O'Reilly, and Declan Murphy. "Sepsis and ‘superbugs’: should we favour the transperineal over the transrectal approach for prostate biopsy?." BJU international 114, no. 3 (2014): 384-388.

6. Liss, Michael A., MAS Behfar Ehdaie, Stacy Loeb, Maxwell V. Meng, Jay D. Raman, Vanessa Spears, CURN Sean P. Stroup et al. "THE PREVENTION AND TREATMENT OF THE MORE COMMON COMPLICATIONS RELATED TO PROSTATE BIOPSY UPDATE." (2016).

7. Briganti, Alberto, Nicola Fossati, James WF Catto, Philip Cornford, Francesco Montorsi, Nicolas Mottet, Manfred Wirth, and Hendrik Van Poppel. "Active surveillance for low-risk prostate cancer: the European Association of Urology position in 2018." European urology 74, no. 3 (2018): 357-368.

8. Klotz, Laurence, Danny Vesprini, Perakaa Sethukavalan, Vibhuti Jethava, Liying Zhang, Suneil Jain, Toshihiro Yamamoto, Alexandre Mamedov, and Andrew Loblaw. "Long-term follow-up of a large active surveillance cohort of patients with prostate cancer." Journal of Clinical Oncology 33, no. 3 (2015): 272-277.

9. Fossati, Nicola, Martina Sofia Rossi, Vito Cucchiara, Giorgio Gandaglia, Paolo Dell’Oglio, Marco Moschini, Nazareno Suardi et al. "Evaluating the effect of time from prostate cancer diagnosis to radical prostatectomy on cancer control: can surgery be postponed safely?." In Urologic Oncology: Seminars and Original Investigations, vol. 35, no. 4, pp. 150-e9. Elsevier, 2017.

10. Wilt, Timothy J., Tien N. Vo, Lisa Langsetmo, Philipp Dahm, Thomas Wheeler, William J. Aronson, Matthew R. Cooperberg, Brent C. Taylor, and Michael K. Brawer. "Radical Prostatectomy or Observation for Clinically Localized Prostate Cancer: Extended Follow-up of the Prostate Cancer Intervention Versus Observation Trial (PIVOT)." European urology (2020).

11. Nacoti, Mirco, Andrea Ciocca, Angelo Giupponi, Pietro Brambillasca, Federico Lussana, Michele Pisano, Giuseppe Goisis et al. "At the epicenter of the Covid-19 pandemic and humanitarian crises in Italy: changing perspectives on preparation and mitigation." NEJM Catalyst Innovations in Care Delivery 1, no. 2 (2020).

Bundle (ABCDE) Checklist for Prevention of CAUTIs

Adherence to infection control principles, standard supplies, procedures and processes

  • Hand hygiene - the most important factor in preventing nosocomial infections
  • Aseptic catheter insertion procedure
  • Proper Foley catheter maintenance, education, and care by nursing staff
  • Foley catheter use surveillance and feedback
Written by: Diane K. Newman, DNP, ANP-BC, FAAN

Risks of Delaying Bladder Cancer Diagnosis- Surveillance and Surgery During COVID-19

The rapid spread of Coronavirus Disease 2019 (COVID-19), caused by the betacoronavirus SARS-CoV-2, throughout the world has had dramatic effects on healthcare systems with impacts far beyond the patients actually infected with COVID-19.

Patients who manifest severe forms of COVID-19 requiring respiratory support typically require this for prolonged durations, with a mean of 13 days of respiratory support reported by the China Medical Treatment Expert Group for COVID-19.1 This lengthy requirement for ventilator support and ICU resources, exacerbated by relatively little excess health system capacity to accommodate epidemics, means that healthcare systems can (and have in the case of many hospitals in Italy) become overwhelmed relatively quickly. In an effort to conserve hospital resources, the American College of Surgeons on March 13th recommended that health systems, hospitals, and surgeons should attempt to minimize, postpone, or outright cancel electively scheduled operations.2 This was done with the primary goal to immediately decrease the use of items essential for the care of patients with COVID-19 including ICU beds, ventilators, personal protective equipment, and terminal cleaning supplies. On March 17th, the American College of Surgeon then provided further guidance on the triage of non-emergent surgeries, including an aggregate assessment of the risk incurred from surgical delays of six to eight weeks or more as compared to the risk (both to the patient and the healthcare system) of proceeding with the operation.3 In the UK, all non-urgent elective surgical procedures have been put on hold for three months to use all of those clinical resources to care for patients with COVID-19.

Most bodies, including the American College of Surgeons, have recommended proceeding with most cancer surgeries. Thus, clinicians and patients must carefully weigh the benefit of proceeding with cancer treatment as scheduled, the risks of COVID-19 to the individual patient, to health care workers caring for patients potentially infected with COVID-19, and the need to conserve health care resources. A severe SARS-CoV-2 phenotype is seen more commonly in men and older, more comorbid patients.4 These characteristics are common in many patients with urologic malignancies, particularly those with bladder cancer. Baseline characteristics among 1,591 patients admitted to the ICU in the Lombardy Region, Italy showed that the median age was 63 years (IQR 56-70), 82% were male, 68% had ≥1 comorbidity, 88% required ventilator support, and the mortality rate was 26%, with a large proportion requiring ongoing ICU level care at the time of data cut-off.5 Work from China demonstrated that patients with cancer had a higher incidence of COVID-19 infection than expected in the general population and had a more severe manifestation of the disease with a significantly higher proportion requiring invasive ventilation in the ICU or death.6 Thus, considering differences in the natural history of different cancers may meaningfully change this balance of risks and benefits.

In the urologic literature, the effect of delays in surgical intervention has been most thoroughly explored in muscle-invasive bladder cancer and prostate cancer. In bladder cancer, these studies have predominately assessed the association between time from transurethral resection of bladder tumor (TURBT) to radical cystectomy. At least nineteen studies have been published assessing this research question. Published October 23, 2019, in European Urology Oncology, Dr. Russell and colleagues provide a contemporary systematic review and meta-analysis of these data.7 The authors undertook a systematic review of Medline®, Embase®, and Ovid® for randomized trials and observational studies assessing the association between delay in treatment and survival (overall or bladder cancer-specific) for patients with bladder cancer. Among 399 identified articles, the authors included 19 studies in systematic review and 10 in meta-analysis. Utilizing the Risk of Bias in Non-randomised studies – of Interventions (ROBINS-I) and the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) criteria, the authors assessed that most studies were of good quality with low or moderate risk of bias. To account for this, the authors performed “leave out one” sensitivity analyses.

Perhaps the most striking conclusion of this systematic review is the considerable heterogeneity in the source literature, including in methodology. There was considerable variation in the nature of the delay investigated: from the diagnosis of bladder cancer to radical cystectomy (10 studies), from TURBT to radical cystectomy (seven studies), from first clinic visit to radical cystectomy (or radiotherapy; one study), from referral to first treatment (one study), and from neoadjuvant chemotherapy to radical cystectomy (four studies). Additionally, the delay interval was operationalized inconsistently across studies with some using time as a continuous variable, some utilizing splines to model a non-linear relationship, and many utilizing a categorical approach with a variety of thresholds.

Among these studies, a number assessed reasons for delay. Identified reasons for delay included scheduling, seeking multiple medical opinions, social issues, misdiagnosis, and patient comorbidity.

Assessing the delay between bladder cancer and survival, four of nine studies found a significant association between delay from diagnosis to radical cystectomy and survival, with a number concluding that tumor stage was a potential confounder in this relationship. Russell and colleagues meta-analyzed three studies with suitable data and found an increased risk of death for patients with significant delays between diagnosis and radical cystectomy (hazard ratio [HR] 1.34, 95% confidence interval [CI] 1.18-1.53; I2=0%).7

Operationalizing delay as the interval between TURBT and radical cystectomy, four of six studies found an association between this time and survival; a meta-analysis of five studies with suitable data for pooling found a borderline non-significant increased risk of overall mortality (odds ratio 1.18, 95% confident interval 0.99-1.41), with significant between study heterogeneity (I2=73%).7 Utilizing a cubic spline to model the non-linear relationship, Kulkarni and colleagues found that the risk of death began to rise beginning at 40 days between TURBT and radical cystectomy.8

An additional five studies assess the association between the time duration between the completion of neoadjuvant chemotherapy and radical cystectomy and survival. Two demonstrated that prolonged durations between neoadjuvant chemotherapy and radical cystectomy was associated with adverse survival outcomes and an additional one demonstrated upstaging was associated with delays. Boeri et al. found that patients who had greater than 10 weeks between the last cycle of neoadjuvant chemotherapy and radical cystectomy had significantly lower cancer-specific and overall survival.9 Chu et al. found similar results10 while three other analyses failed to support these results. A meta-analysis of three studies with data suitable for pooling failed to demonstrate a significant association between delays from the end of neoadjuvant chemotherapy to radical cystectomy with survival (HR 1.04, 95% CI 0.93-1.16; I2=82%).7

Particularly relevant in the context of the COVID-19 pandemic, Audenet and colleagues found that delays to neoadjuvant chemotherapy of greater than eight weeks were associated with an increased risk of upstaging, while they found no harm in delays up to six months from diagnosis to radical cystectomy, assuming that neoadjuvant chemotherapy was administered in the meantime.11

While the meta-analysis of Russell and colleagues focused on patients with urothelial histology, recently Lin-Brande examined outcomes for patients with variant histology undergoing radical cystectomy.12 In this analysis, patients with variant histology had a similar time from diagnosis to radical cystectomy as those with urothelial histology. In this cohort, delays from diagnosis to radical cystectomy were associated with worse overall survival (HR 1.36, 95% CI 1.11-1.65 per month of delay) after adjusting for relevant clinicopathologic features. The authors then subsequently dichotomized surgical delays using thresholds of four-, eight-, and 12-weeks. On multivariable analysis, no difference in overall survival was apparent when “early” versus “late” was dichotomized at four weeks (hazard ratio 0.92, 95% CI 0.32-2.59) or eight weeks (HR 1.50, 95% CI 0.68-3.29) but significant differences were apparent when delayed surgery was defined as that beyond 12 weeks following diagnosis (HR 3.45, 95% CI 1.51-7.86).

There is somewhat less evidence in patients with non-muscle invasive disease, with significant differences between patients with low-grade and high-grade disease. Low-grade non-muscle invasive bladder cancer has low cancer-specific mortality13 and there is no evidence of harm from delays in management.14-16

In contrast, for patients with high-grade non-muscle invasive disease, progression to muscle invasion/metastases occurs in 15-40% and 10-20% of patients may die from bladder cancer.17,18 In patients who underwent radical cystectomy for recurrent non-muscle invasive bladder cancer following Bacillus Calmette-Guerin (BCG) with or without further intravesical therapy, the delay caused by an additional (unsuccessful) course of intravesical therapy did not result in differences in five-year overall or cancer-specific survival despite a median delay of 1.7 years.19 In contrast, among patients with non-muscle invasive (cT1) micropapillary bladder cancer treated with upfront radical cystectomy or intravesical BCG, Willis et al. demonstrated significantly poorer survival outcomes for patients who underwent initial intravesical therapy.20

In the absence of data, guidance on the care of patients with high-grade non-muscle invasive bladder cancer has relied upon expert guidance. A collaborative review pre-published in European Urology suggested that these patients should receive induction BCG and at least one course of maintenance therapy as the first-line treatment. The authors recommended that re-resection should be continued for patients with pT1 disease while this may be omitted for those with pTa and muscle in the initial resection.

The data regarding delays to radical cystectomy demonstrate considerable heterogeneity with mixed results. This is in large part due to varying definitions of a delay, despite the threshold of 12 weeks advocated in EAU guidelines.21 However, in aggregate, these data suggest that prolonged delays (likely 90 days although potentially as short as 40 days) between bladder cancer diagnosis or TURBT and radical cystectomy are associated with worse survival. However, the data are mixed and pooled results demonstrate considerable heterogeneity. Further, when neoadjuvant chemotherapy is employed, delays to radical cystectomy no longer appear to be significant. Finally, an analysis of funnel plots indicates that there is a publication bias towards studies which demonstrate worse survival associated with delays suggesting that this finding may be exaggerated in the literature.7

A recent multi-institutional analysis from Campi and colleagues assessed the proportion of patients undergoing urologic oncology surgery who had “nondeferrable” indications for treatment.22 The authors identified 2387 patients undergoing radical cystectomy, radical nephroureterectomy, nephrectomy, and radical prostatectomy in the past 12 months at San Luigi Hospital in Turin, San Raffaele Hospital in Milan, and Careggi Hospital in Florence. Assessing patients undergoing radical cystectomy, the authors considered all cases “nondeferrable”. Radical cystectomy comprised 36.2% of all so-called high-priority or nondeferrable cases. They also noted that a large proportion of these patients (50%) are at elevated perioperative risk (defined at American Society of Anesthesiologists score ≥ 3).

In the context of non-muscle invasive disease, delays appear to be significantly more likely to cause harm in patients with high-grade than low-grade disease. However, the evidence base in non-muscle invasive disease is much weaker than for muscle-invasive bladder cancer.

However, when considering delays in performing TURBT, it is not always apparent whether the patient has non-muscle invasive or muscle-invasive disease. Wallace and colleagues examined delays throughout the trajectory of patients with a new diagnosis of urothelial carcinoma, including a preponderance of non-muscle invasive disease (pTa = 51% and pT1 = 22%).23 The authors divided delays in the evaluation of these patients into three: from initial symptom onset to general practitioner presentation (patient-derived delay), from general practitioner presentation to hospital referral (general practitioner-derived delay), and from hospital referral to TURBT (hospital-derived delay). Shorter delays from initial symptom onset to general practitioner presentation were associated with lower tumor stage and improved five-year survival. In this analysis, the authors dichotomized this patient-derived delay at 14 days. Notably, patients with shorter general practitioner-derived delay had worse survival, likely reflecting a selection bias in which patients with particularly worrisome presentations received expedited care. Finally, hospital-derived delays were not significantly associated with survival, whether adjusted for tumor stage or not. When considered in aggregate, total delays from initial symptom onset to TURBT were not significantly associated with survival.

During the COVID-19 pandemic, there will be regional variations in the risk of infection and availability of resources, both with regards to medical treatment and operating room availability. Expert recommendations from the pre-published paper in European Urology suggest it is reasonable that patients with high-grade NMIBC should undergo induction BCG and maintenance therapy if possible. For high-risk NMIBC, radical cystectomy should still be offered if the resources are feasible and the patient’s comorbidity profile does not place them at higher post-operative COVID-19 risk. Based on the available literature, delays in radical cystectomy of up to three months may be safe for muscle-invasive bladder cancer. However, clinicians should prioritize high-grade bladder cancer (NMIBC and muscle-invasive) over other urologic oncology procedures during COVID-19 restrictions.

Written by: Zachary Klaassen, MD, MSc, Assistant Professor of Urology, Georgia Cancer Center, Augusta University/Medical College of Georgia, Atlanta, Georgia

Published Date: April 20th, 2020

Written by: Zachary Klaassen, MD, MSc
References: 1. Guan, Wei-jie, Zheng-yi Ni, Yu Hu, Wen-hua Liang, Chun-quan Ou, Jian-xing He, Lei Liu et al. "Clinical characteristics of coronavirus disease 2019 in China." New England Journal of Medicine (2020).

2. March 13, Online, and 2020. “COVID-19: Recommendations for Management of Elective Surgical Procedures.” American College of Surgeons. Accessed April 10, 2020. https://www.facs.org/covid-19/clinical-guidance/elective-surgery.

3.  March 17, Online, and 2020. “COVID-19: Guidance for Triage of Non-Emergent Surgical Procedures.” American College of Surgeons. Accessed April 17, 2020. https://www.facs.org/covid-19/clinical-guidance/triage.

4. COVID, CDC, and Response Team. "Severe outcomes among patients with coronavirus disease 2019 (COVID-19)—United States, February 12–March 16, 2020." MMWR Morb Mortal Wkly Rep 69, no. 12 (2020): 343-346.

5. Grasselli, Giacomo, Alberto Zangrillo, Alberto Zanella, Massimo Antonelli, Luca Cabrini, Antonio Castelli, Danilo Cereda et al. "Baseline Characteristics and Outcomes of 1591 Patients Infected With SARS-CoV-2 Admitted to ICUs of the Lombardy Region, Italy." JAMA (2020).

6. Liang, Wenhua, Weijie Guan, Ruchong Chen, Wei Wang, Jianfu Li, Ke Xu, Caichen Li et al. "Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China." The Lancet Oncology 21, no. 3 (2020): 335-337.

7. Russell, Beth, Fredrik Liedberg, Muhammad Shamim Khan, Rajesh Nair, Ramesh Thurairaja, Sachin Malde, Pardeep Kumar, Richard T. Bryan, and Mieke Van Hemelrijck. "A Systematic Review and Meta-analysis of Delay in Radical Cystectomy and the Effect on Survival in Bladder Cancer Patients." European urology oncology (2019).

8. Kulkarni, Girish S., David R. Urbach, Peter C. Austin, Neil E. Fleshner, and Andreas Laupacis. "Longer wait times increase overall mortality in patients with bladder cancer." The Journal of urology 182, no. 4 (2009): 1318-1324.

9. Boeri, Luca, Matteo Soligo, Igor Frank, Stephen A. Boorjian, R. Houston Thompson, Matthew Tollefson, Fernando J. Quevedo, John C. Cheville, and R. Jeffrey Karnes. "Delaying radical cystectomy after neoadjuvant chemotherapy for muscle-invasive bladder cancer is associated with adverse survival outcomes." European urology oncology 2, no. 4 (2019): 390-396.

10. Chu, Alice T., Sarah K. Holt, Jonathan L. Wright, Jorge D. Ramos, Petros Grivas, Evan Y. Yu, and John L. Gore. "Delays in radical cystectomy for muscle‐invasive bladder cancer." Cancer 125, no. 12 (2019): 2011-2017.

11. Audenet, François, John P. Sfakianos, Nikhil Waingankar, Nora H. Ruel, Matthew D. Galsky, Bertram E. Yuh, and Greg E. Gin. "A delay≥ 8 weeks to neoadjuvant chemotherapy before radical cystectomy increases the risk of upstaging." In Urologic Oncology: Seminars and Original Investigations, vol. 37, no. 2, pp. 116-122. Elsevier, 2019.

12. Lin-Brande, Michael, Shane M. Pearce, Akbar N. Ashrafi, Azadeh Nazemi, Madeleine L. Burg, Saum Ghodoussipour, Gus Miranda, Hooman Djaladat, Anne Schuckman, and Siamak Daneshmand. "Assessing the Impact of Time to Cystectomy for Variant Histology of Urothelial Bladder Cancer." Urology 133 (2019): 157-163.

13. Lopez-Beltran, Antonio, and Rodolfo Montironi. "Non-invasive urothelial neoplasms: according to the most recent WHO classification." European urology 46, no. 2 (2004): 170-176.

14. Soloway, Mark S., Darren S. Bruck, and Sandy S. Kim. "Expectant management of small, recurrent, noninvasive papillary bladder tumors." The Journal of urology 170, no. 2 (2003): 438-441.

15. Guidance, N. I. C. E. "Bladder cancer: diagnosis and management of bladder cancer." BJU Int 120, no. 6 (2017): 755-765.

16. Matulay, Justin T., Mark Soloway, J. Alfred Witjes, Roger Buckley, Raj Persad, Donald L. Lamm, Andreas Boehle et al. "Risk‐adapted management of low‐grade bladder tumours: recommendations from the International Bladder Cancer Group (IBCG)." BJU international 125, no. 4 (2020): 497-505.

17. Klaassen, Zachary, Ashish M. Kamat, Wassim Kassouf, Paolo Gontero, Humberto Villavicencio, Joaquim Bellmunt, Bas WG van Rhijn, Arndt Hartmann, James WF Catto, and Girish S. Kulkarni. "Treatment strategy for newly diagnosed T1 high-grade bladder urothelial carcinoma: new insights and updated recommendations." European urology 74, no. 5 (2018): 597-608.

18. Thomas, Francis, Aidan P. Noon, Naomi Rubin, John R. Goepel, and James WF Catto. "Comparative outcomes of primary, recurrent, and progressive high-risk non–muscle-invasive bladder cancer." European urology 63, no. 1 (2013): 145-154.

19. Haas, Christopher R., LaMont J. Barlow, Gina M. Badalato, G. Joel DeCastro, Mitchell C. Benson, and James M. McKiernan. "The timing of radical cystectomy for bacillus Calmette-Guerin failure: comparison of outcomes and risk factors for prognosis." The Journal of urology 195, no. 6 (2016): 1704-1709.

20. Willis, Daniel L., Mario I. Fernandez, Rian J. Dickstein, Sahil Parikh, Jay B. Shah, Louis L. Pisters, Charles C. Guo et al. "Clinical outcomes of cT1 micropapillary bladder cancer." The Journal of urology 193, no. 4 (2015): 1129-1134.

21. Witjes, J. Alfred, Thierry Lebret, Eva M. Compérat, Nigel C. Cowan, Maria De Santis, Harman Maxim Bruins, Virginia Hernandez et al. "Updated 2016 EAU guidelines on muscle-invasive and metastatic bladder cancer." European urology 71, no. 3 (2017): 462-475.

22. Campi, Riccardo, Daniele Amparore, Umberto Capitanio, Enrico Checcucci, Andrea Salonia, Cristian Fiori, Andrea Minervini et al. "Assessing the Burden of Nondeferrable Major Uro-oncologic Surgery to Guide Prioritisation Strategies During the COVID-19 Pandemic: Insights from Three Italian High-volume Referral Centres." European Urology (2020).

23. Wallace, D. M. A., R. T. Bryan, J. A. Dunn, G. Begum, S. Bathers, and West Midlands Urological Research Group. "Delay and survival in bladder cancer." BJU international 89, no. 9 (2002): 868-878.

Beyond Bladder Cancer: Bacillus Calmette-Guérin (BCG) Vaccination Revisited as a Strategy to Reduce COVID-19 Related Adverse Events in High Risk Health Care Workers and the Elderly

First Published April 2, 2020

The ongoing pandemic involving severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its resulting coronavirus disease 2019 (COVID-19) has caused widespread infection worldwide, with over 660,000 confirmed cases as of March 28, 2020 and nearly 31,000 deaths.1 Data from the Italian National Institute of Health (Istituto Superiore di Sanità [ISS]) where fatalities are thus far the highest suggest a fatality rate of 7.2%, significantly higher than that which has been observed in other countries.2 Elderly patients are at greatest risk of mortality from COVID-19, and approximately 23% of the Italian populace is aged 65 years or older, making the country particularly vulnerable.2

Written by: Vikram M. Narayan, Paul Hegarty, Gianluca Giannarini, Rick Bangs, Stephanie Chisolm, and Ashish M. Kamat
References:

1. University JH. Johns Hopkins Center for Systems Science and Engineering Coronavirus Resource Center n.d.

2. Onder G, Rezza G, Brusaferro S. Case-Fatality Rate and Characteristics of Patients Dying in Relation to COVID-19 in Italy. JAMA 2020. doi:10.1001/jama.2020.4683.

3. Liang W, Guan W, Chen R, Wang W, Li J, Xu K, et al. Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China. Lancet Oncol 2020;21:335–7. doi:10.1016/S1470-2045(20)30096-6.

4. Moulton LH, Rahmathullah L, Halsey NA, Thulasiraj RD, Katz J, Tielsch JM. Evaluation of non-specific effects of infant immunizations on early infant mortality in a southern Indian population. Trop Med Int Heal 2005;10:947–55. doi:10.1111/j.1365-3156.2005.01434.x.

5. Aaby P, Roth A, Ravn H, Napirna BM, Rodrigues A, Lisse IM, et al. Randomized trial of BCG vaccination at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J Infect Dis 2011;204:245–52. doi:10.1093/infdis/jir240.

6. Leentjens J, Kox M, Stokman R, Gerretsen J, Diavatopoulos DA, van Crevel R, et al. BCG Vaccination Enhances the Immunogenicity of Subsequent Influenza Vaccination in Healthy Volunteers: A Randomized, Placebo-Controlled Pilot Study. J Infect Dis 2015;212:1930–8. doi:10.1093/infdis/jiv332.

7. Arts RJW, Moorlag SJCFM, Novakovic B, Li Y, Wang S-Y, Oosting M, et al. BCG Vaccination Protects against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with Trained Immunity. Cell Host Microbe 2018;23:89-100.e5. doi:10.1016/j.chom.2017.12.010.

8. Hegarty PK, Sfakianos JP, Giannarini G, DiNardo AR, Kamat AM. Coronavirus disease 2019 (Covid-19) and Bacillus Calmette-Guérin (BCG): what is the link? Eur Urol Oncol 2020 in press

Definition – External Urine Collection Device

An external urine collection device (EUCD) is defined as a catheter or product that attaches to the perineum. These collection systems drain urine via tubing attached to a bag or via tubing that suctions urine to a container. EUCDs are primarily used in men or women with urinary incontinence. They are either one-time disposable devices or reused multiple times and are made from many materials but the most common material is latex and silicone.
Written by: Diane K. Newman, DNP, ANP-BC, FAAN
References: 1. Beeson, Terrie, and Carmen Davis. "Urinary management with an external female collection device." Journal of Wound, Ostomy, and Continence Nursing 45, no. 2 (2018): 187.
2. Deng, Donna Y. "Urologic Devices." In Clinical Application of Urologic Catheters, Devices and Products, pp. 173-220. Springer, Cham, 2018.
3. Eckert, Lorena, Lisa Mattia, Shilla Patel, Rowena Okumura, Priscilla Reynolds, and Ingrid Stuiver. "Reducing the Risk of Indwelling Catheter–Associated Urinary Tract Infection in Female Patients by Implementing an Alternative Female External Urinary Collection Device: A Quality Improvement Project." Journal of Wound Ostomy & Continence Nursing 47, no. 1 (2020): 50-53.
4. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.
5.Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
6. Fader, Mandy, Donna Bliss, Alan Cottenden, Katherine Moore, and Christine Norton. "Continence products: research priorities to improve the lives of people with urinary and/or fecal leakage." Neurourology and Urodynamics: Official Journal of the International Continence Society 29, no. 4 (2010): 640-644.
7. Geng, V., H. Cobussen-Boekhorst, H. Lurvink, I. Pearce, and S. Vahr. "Evidence-based guidelines for best practice in urological health care: male external catheters in adults urinary catheter management." Arnhem: European Association of Urology Nurses (2016).
8. Gray, Mikel, Claudia Skinner, and Wendy Kaler. "External collection devices as an alternative to the indwelling urinary catheter: evidence-based review and expert clinical panel deliberations." Journal of Wound, Ostomy, and Continence Nursing 43, no. 3 (2016): 301.
9. Lachance, Chantelle C., and Aleksandra Grobelna. "Management of Patients with Long-Term Indwelling Urinary Catheters: A Review of Guidelines." (2019).
10. Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
11. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.

Safe and Efficacious Therapies Urgently Needed for the Difficult to Treat Non-muscle Invasive Bladder Cancer Patient Population

A newly published systematic review and meta-analysis: Evidence-based Assessment of Current and Emerging Bladder-sparing Therapies for Non–muscle-invasive Bladder Cancer After Bacillus Calmette-Guerin Therapy: A Systematic Review and Meta-analysis 

Written by: Catherine Ryan

Types and Materials – External Urine Collection Devices

The shape and material of external urine collection devices (EUCD) have changed over the past 20 years. Historically, most EUCDs were made from latex that allowed for flexibility but also increased the risk of an allergic reaction. Latex-based sheath devices are still available but more recent ones are constructed from non-allergenic silicone. Most EUCDs are open at the distal end (tip) allowing urine to drain through attached tubing connected to a drainage bag. 

figure-1-materials2x_1.jpg

There are two broad categories, those that are single-use disposable products (in-place for only one to two days), and those that are reusable for multiple times.

If the EUCD has adhesive, prior to its application, the skin should be cleansed and pubic hair at the base of the penis in men and the perineum in women should be removed. The hair should be trimmed, not shaved, because shaving causes more irritation. The EUCD can be removed by loosening the adhesive with a warm, wet cloth. 

We are categorizing the types of EUCDs as follows:

table 1 external urine collection devices2x 1

Written by: Diane K. Newman, DNP, ANP-BC, FAAN, Adjunct Professor of Urology in Surgery, Research Investigator Senior, Perelman School of Medicine, Co-Director, Penn Center for Continence and Pelvic Health, Division of Urology, University of Pennsylvania, Philadelphia, Pennsylvania

April 2020

Written by: Diane K. Newman, DNP, ANP-BC, FAAN
References: 1. Beeson, Terrie, and Carmen Davis. "Urinary management with an external female collection device." Journal of Wound, Ostomy, and Continence Nursing 45, no. 2 (2018): 187.
2. Cottenden, Alan, D. Z. Bliss, B. Buckley, M. Fader, C. Gartley, D. Hayder, J. Ostaszkiewicz, and M. Wilde. "Management using continence products." Incontinence (2013): 1651-1786.
3. Doherty, Willie. "The InCare Retracted Penis Pouch: an alternative for incontinent men." British journal of nursing 11, no. 11 (2002): 781-784.
4. Gray, Mikel, Claudia Skinner, and Wendy Kaler. "External collection devices as an alternative to the indwelling urinary catheter: evidence-based review and expert clinical panel deliberations." Journal of Wound, Ostomy, and Continence Nursing 43, no. 3 (2016): 301.
5. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.
6. Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
7. Newman, D. K., and A. J. Wein. "Managing and treating urinary incontinence. 2009 Baltimore."
8. Newman, Diane K. "Internal and external urinary catheters: a primer for clinical practice." Ostomy/wound management 54, no. 12 (2008): 18-35.
9. Newman, Diane K. "Incontinence products and devices for the elderly." Urologic nursing 24, no. 4 (2004): 316-333.
10. Newman, Diane K., Mandy Fader, and Donna Z. Bliss. "Managing incontinence using technology, devices, and products: directions for research." Nursing Research 53, no. 6S (2004): S42-S48.
11. Newman, D. K. "The use of devices and products." American Journal of Nursing 3 (2003): 50-51.
12. Pomfret, I. "Penile sheaths: a guide to selection and fitting." Journal of Community Nursing 20, no. 11 (2006): 14.
13. Smart, Clare. "Male urinary incontinence and the urinary sheath." British Journal of Nursing 23, no. Sup9 (2014): S20-S25.
14. Wells, Mandy. "Managing urinary incontinence with BioDerm® external continence device." British Journal of Nursing 17, no. Sup4 (2008): S24-S29.
15. Vaidyanathan, S., B. M. Soni, G. Singh, P. Sett, E. Brown, and S. Markey. "Possible use of BioDerm External Continence Device in selected, adult, male spinal cord injury patients." Spinal cord 43, no. 4 (2005): 260-261.

The History of Imaging for Prostate Cancer

Diagnosis and assessment of primary tumor – TRUS and mpMRI

Historically, prostate cancer diagnosis was made on the basis of transrectal or transperineal needle biopsy guided by digital palpation per rectum (so-called, finger guided biopsies).1 These biopsies were typically directed at palpable abnormalities. A number of significant changes occurred to this approach beginning in the early 1990s. First, a systematic approach to prostate biopsy advocated by Hodge et al., as opposed to directed cores, was widely adopted.2

Second, the use of transrectal ultrasound (TRUS) for prostate visualization and biopsy guidance became widespread. The use of TRUS allowed for direct visualization of the prostate, any of its anomalies, as well as the biopsy needle. Thus, TRUS-guided prostate biopsy became the gold standard approach to prostate cancer diagnosis.3 However, there are well-known limitations to TRUS-guided prostate biopsy including inherent random and systematic errors. Unless clear visible hypoechoic suspicious areas are seen in TRUS, sampling occurs by chance, and specific zones are under-sampled, including the anterior region and apex.4 Further, TRUS-guided systematic prostate biopsy can miss up to 20% of clinically significant prostate cancer, resulting in underdiagnosis and undertreatment.5 However, at the same time, TRUS-guided systematic prostate biopsy detects a relatively high percentage of clinically insignificant prostate cancer (Gleason grade group [GGG] 1), which may result in overtreatment.6

Thus, thirdly, multiparametric magnetic resonance has recently been evaluated for the identification of prostate lesions likely to be cancerous, as well as the guidance of prostate biopsy.

Initially, MRI was used as a staging test in patients with prostate cancer for assessment of direct extra-prostatic extension utilizing T2-weighted imaging. This approach was marked by significant variability in diagnostic performance, limited ability to detect microscopic disease and inability to localize the tumor within the gland itself.7 These factors limited the widespread adoption of MRI for local tumor staging. Indeed, to this data, TNM staging for prostate cancer relies on digital rectal examination rather than radiographic findings for local tumor staging.

However, multiparametric MRI, particularly with the addition of diffusion-weighted imaging has allowed for increasingly informative studies, including the visualization of tumors within the prostate. This has allowed for the use of mpMRI to guide prostate biopsy, either directly with in-bore biopsy or more commonly using a fusion device platform.8 When performed in the evaluation of patients with elevated prostate-specific antigen (PSA) levels with previous negative prostate biopsy, multi-parametric magnetic resonance imaging has been shown to identify clinically significant prostate cancers which would have been otherwise missed by routine systematic biopsy.9 A recent systematic review and meta-analysis from Kasivisvanathan and colleagues suggested that multi-parametric magnetic resonance imaging targeted biopsy detects more clinically significant prostate cancer than standard TRUS-guided systematic biopsy alone and requires fewer prostate cores to do so; that the question of whether to include systematic biopsy along with multi-parametric magnetic resonance imaging targeted biopsy remains controversial; and that the omission of the systematic biopsy risks missing the diagnosis of clinically significant disease in approximately 13% of men while the inclusion of systematic biopsy increases the likelihood of diagnosing clinically insignificant prostate cancer.10

The most recent European Association of Urology Prostate cancer guidelines conclude that, when at least one functional imaging technique is employed, mpMRI has good sensitivity for the detection and localization of clinically significant (Gleason Grade Group 2 or greater) prostate cancers6 with lower sensitivity for the detection of Gleason Grade Group 1 cancers, likely a beneficial characteristic. Potential limitations of the widespread use of a multi-parametric magnetic resonance imaging driven diagnostic pathway include only a moderate inter-reader reproducibility of multi-parametric magnetic resonance imaging, the lack of standardization of targeted biopsy, and cost-effectiveness concerns in certain jurisdictions.

Even more recently, high-resolution micro-ultrasound has emerged as a novel imaging modality for prostate cancer. High-resolution micro-ultrasound has a very fine resolution (approximately 70 µm) which allows for visualization of alterations in ductal anatomy and cellular density consistent with prostate tumors.11 In early experiences, high-resolution micro-ultrasound has demonstrated an ability to detect clinically significant cancers that were not apparent on either traditional TRUS or mpMRI.12 In contrast to mpMRI, high-resolution micro-ultrasound has the advantage of providing real-time imaging results, a finding that authors from the Cleveland Clinic demonstrated was associated with a relative increase in prostate cancer detection of 26.7%.12 Aggregate data from early clinical experience at multiple centers suggests that high-resolution micro-ultrasound has comparable or increased sensitivity for clinically significant prostate cancer compared with mpMRI and comparable or slightly reduced specificity.11

Distant staging – from radiographs to molecularly targeted imaging

While mpMRI has revolutionized imaging of the prostate and substantially changed the diagnostic algorithm for prostate cancer, perhaps even greater changes have occurred in the imaging for distant disease.

Initially, a radiographic diagnosis of bony prostate cancer metastasis was made on the basis of plain radiographs. However, bony metastases may be difficult to identify based on plain films as an extensive bone mineral loss (exceeding 30-50%) may be required before such changes are radiographically apparent.13 However, plain films remain useful for the immediate investigation of patients who present with bony pain and for the assessment of bony stability in those deemed at risk of pathologic fracture.

Following plain projectional radiography, skeletal scintigraphy was the next imaging modality widely adopted for the assessment of bony metastases in patients with prostate cancer. To date, it remains widely utilized and is currently recommended, along with abdominal and pelvic computed tomography, for the staging of patients according to many guideline bodies. Skeletal scintigraphy, when performed in patients with known cancer in the absence of bony pain, has a sensitivity of 86% and specificity of 81% for the detection of metastatic lesions.13 As with any imaging modality, these characteristics differ somewhat on the basis of the patient population being tested (i.e. the pre-test probability or population-based disease prevalence). Among patients with prostate cancer, PSA levels are predictive of the likelihood of a positive bone scan. Across a number of different cancers, Yang et al. found that bone scintigraphy had a specificity of 81.4% and sensitivity of 86.0%, on a per-patient basis, for the detection of bony metastases.14

Computed tomography has been utilized for the assessment of nodal metastatic disease, visceral disease, and bony metastasis. CT is highly sensitive for both osteoblastic tumors (such as prostate cancer) and osteolytic lesions in the cortical bone but is less sensitive in tumors that are restricted to the marrow space.13 As a result, CT is of relatively limited utility as a screening test for bony metastasis due to relatively low sensitivity (73%) despite excellent specificity (95%) – numbers based on a large scale meta-analysis from Yang and colleagues.14 For this reason, conventional staging recommendations for patients with prostate cancer include bony scintigraphy for the detection of bony lesions along with computed tomography for identification of nodal/visceral lesions and correlation of any bony lesions.15

In addition to its role in the local staging of the prostate and guidance of prostate biopsy, mpMRI may also assist with evaluation for distant metastatic disease. Routine pelvic/prostate MRI typically allows for assessment of local/regional nodal involvement including obturator and external iliac nodal chains. However, the high soft-tissue contrast and high spatial resolution afforded by MRI call also allow for the identification of bony metastasis in marrow spaces much early than would be apparent based on CT scan.14 Further, use of T1-weighted sequences and STIR sequences can allow for adequate assessment for bony metastasis without the need for intravenous contrast agents; use of MRI for staging does not require the use of ionizing radiation. Thus, abdominal/pelvic or whole-body MRI may be considered for the identification of distant metastatic disease. Additionally, MRI with contrast has become the imaging modality of choice for the evaluation of liver metastases.16 Thus, this approach may be particularly valuable in patients at a high risk of visceral metastatic disease.

Traditional positron emission tomography (PET) imaging utilizing fluorodeoxyglucose (FDG) is not typically effective in the initial diagnosis of prostate cancer metastasis owing to the relatively low metabolic activity associated with the disease. However, at least four other PET imaging approaches have been assessed and employed in patients with prostate cancer including 18F-NaF PET/CT, choline-based PET/CT, fluciclovine (Axumin®) PET/CT, and PSMA-targeted PET/CT.17 These modalities have been used in the staging of both primary and recurrent prostate cancer. While clearly improved compared to bony scintigraphy, the limitations are similar – namely, that sensitivity is highly dependent on PSA levels. However, choline-based PET/CT has demonstrated significantly higher sensitivity for the diagnosis of metastatic lesions at the time of biochemical recurrence compared to conventional imaging with a bone scan and computed tomography.17 However, compared to MRI, the benefits of choline-based PET/CT are less clear.18 MRI clearly outperformed choline-based PET/CT for the detection of local recurrence (36.1% vs 1.6%), while choline-PET/CT was superior for identification of lymph node metastasis and both were effective at identifying bony metastatic disease.19

Choline-based PET/CT is not widely available in the United States. However, fluciclovine PET/CT (also known as Axumin® PET/CT) which utilizes the proliferation of tumor cells for localization, is much more available. Fluciclovine (18F-FACBC; 1-amino-3-fluorine 18F-flurocyclobutane-1-carboxylic acid) is a synthetic amino acid analog with the advantage of negligible renal uptake and no activity in the urinary tract.18 Nevertheless, non-specific prostate uptake limits its utility in the identification of primary prostate tumors due to an inability to distinguish from benign prostatic inflammation. Instead, fluciclovine-PET/CT has proven efficacy in the detection of recurrent prostate cancer with biochemical recurrence following local therapy, with a sensitivity of 90% and specificity of 40% (higher in distant, 97%, and nodal disease, 55%, than locally).20 Compared to choline-PET/CT, fluciclovine-PET/CT demonstrated lower false-negatives and false-positive rates in patients with biochemical recurrence.21, 22

Finally, receptor-targeted PET imaging has recently been examined, most notably, PSMA-based PET/CT. PSMA is a transmembrane glycoprotein found on prostatic epithelium. The ratio of PSMA to its truncated isoform (PSM’) is proportional to tumor aggressivity. The most well examined PSMA based approach is 68Ga-PSMA-PET/CT. In patients with biochemical recurrence following radical prostatectomy, 68Ga-PSMA-PET/CT demonstrated superior detection rates of metastatic disease (56%) compared with fluciclovine-PET/CT (13%).23 This benefit was consistent in detecting pelvic nodal disease and extrapelvic disease. PSMA-based PET/CT demonstrated a particular benefit in the evaluation of patients with low absolute PSA levels. Further, 68Ga-PSMA-PET/CT appears to be superior to MRI in primary staging of patients prior to local therapy.24 Other radiotracers including 18F-DCFPyL and 177Lu-PSMA-617 have recently been examined in place of 68Ga-PSMA.25

Recent work has also assessed the role of PET/MRI, rather than PET/CT. This approach leverages the advantages of the sensitivity of receptor-targeted imaging and the spatial resolution of MRI.24

Conclusion

The evolution of imaging in prostate cancer has allowed a more nuanced understanding of the disease. Assessing the local tumor, both mpMRI and high-resolution micro-ultrasound allow for a more informed prostate biopsy which may assist in more accurate initial disease characterization26 as well as local staging. Ongoing advances in receptor-targeted PET imaging continue to refine the identification of metastatic disease. This has important implications for what we understand to be M0 and M1 prostate cancer. Whether early detection of metastatic disease utilizing these modalities translates into improvements in patient outcomes, or simply lead-time bias, remains to be assessed.

Published Date: March 2020
Written by: Zachary Klaassen, MD, MSc
References: 1. Shinohara, K., V. A. Master, T. Chi, and P. R. Carroll. "Prostate needle biopsy techniques and interpretation." Genitourinary Oncology. Philadelphia, Lippincott, Williams & Wilkins (2006): 111-119.
2. Hodge, Kathryn K., John E. McNeal, Martha K. Terris, and Thomas A. Stamey. "Random systematic versus directed ultrasound guided transrectal core biopsies of the prostate." The Journal of urology 142, no. 1 (1989): 71-74.
3. Heidenreich, Axel, Patrick J. Bastian, Joaquim Bellmunt, Michel Bolla, Steven Joniau, Theodor van der Kwast, Malcolm Mason et al. "EAU guidelines on prostate cancer. Part 1: screening, diagnosis, and local treatment with curative intent—update 2013." European urology 65, no. 1 (2014): 124-137.
4. Kongnyuy, Michael, Abhinav Sidana, Arvin K. George, Akhil Muthigi, Amogh Iyer, Michele Fascelli, Meet Kadakia et al. "The significance of anterior prostate lesions on multiparametric magnetic resonance imaging in African-American men." In Urologic Oncology: Seminars and Original Investigations, vol. 34, no. 6, pp. 254-e15. Elsevier, 2016.
5. Schouten, Martijn G., Marloes van der Leest, Morgan Pokorny, Martijn Hoogenboom, Jelle O. Barentsz, Les C. Thompson, and Jurgen J. Fütterer. "Why and where do we miss significant prostate cancer with multi-parametric magnetic resonance imaging followed by magnetic resonance-guided and transrectal ultrasound-guided biopsy in biopsy-naïve men?." European urology 71, no. 6 (2017): 896-903.
6. Mottet, Nicolas, Joaquim Bellmunt, Michel Bolla, Erik Briers, Marcus G. Cumberbatch, Maria De Santis, Nicola Fossati et al. "EAU-ESTRO-SIOG guidelines on prostate cancer. Part 1: screening, diagnosis, and local treatment with curative intent." European urology 71, no. 4 (2017): 618-629.
7. Rifkin, Matthew D., Elias A. Zerhouni, Constantine A. Gatsonis, Leslie E. Quint, David M. Paushter, Jonathan I. Epstein, Ulrike Hamper, Patrick C. Walsh, and Barbara J. McNeil. "Comparison of magnetic resonance imaging and ultrasonography in staging early prostate cancer: results of a multi-institutional cooperative trial." New England Journal of Medicine 323, no. 10 (1990): 621-626.
8. Siddiqui, M. Minhaj, Soroush Rais-Bahrami, Baris Turkbey, Arvin K. George, Jason Rothwax, Nabeel Shakir, Chinonyerem Okoro et al. "Comparison of MR/ultrasound fusion–guided biopsy with ultrasound-guided biopsy for the diagnosis of prostate cancer." Jama 313, no. 4 (2015): 390-397.
9. Vourganti, Srinivas, Ardeshir Rastinehad, Nitin K. Yerram, Jeffrey Nix, Dmitry Volkin, An Hoang, Baris Turkbey et al. "Multiparametric magnetic resonance imaging and ultrasound fusion biopsy detect prostate cancer in patients with prior negative transrectal ultrasound biopsies." The Journal of urology 188, no. 6 (2012): 2152-2157.
10. Kasivisvanathan, Veeru, Armando Stabile, Joana B. Neves, Francesco Giganti, Massimo Valerio, Yaalini Shanmugabavan, Keiran D. Clement et al. "Magnetic resonance imaging-targeted biopsy versus systematic biopsy in the detection of prostate cancer: a systematic review and meta-analysis." European urology (2019).
11. Klotz, CM Laurence. "Can high resolution micro-ultrasound replace MRI in the diagnosis of prostate cancer?." European urology focus (2019).
12. Abouassaly, Robert, Eric A. Klein, Ahmed El-Shefai, and Andrew Stephenson. "Impact of using 29 MHz high-resolution micro-ultrasound in real-time targeting of transrectal prostate biopsies: initial experience." World journal of urology (2019): 1-6.
13. Heindel, Walter, Raphael Gübitz, Volker Vieth, Matthias Weckesser, Otmar Schober, and Michael Schäfers. "The diagnostic imaging of bone metastases." Deutsches Ärzteblatt International 111, no. 44 (2014): 741.
14. Yang, Hui-Lin, Tao Liu, Xi-Ming Wang, Yong Xu, and Sheng-Ming Deng. "Diagnosis of bone metastases: a meta-analysis comparing 18 FDG PET, CT, MRI and bone scintigraphy." European radiology 21, no. 12 (2011): 2604-2617.
15. Network NCC. NCCN Clinical Practice Guideslines in Oncology: Prostate Cancer - Version 1.2019. 2019.
16. Namasivayam, Saravanan, Diego R. Martin, and Sanjay Saini. "Imaging of liver metastases: MRI." Cancer Imaging 7, no. 1 (2007): 2.
17. Li, Roger, Gregory C. Ravizzini, Michael A. Gorin, Tobias Maurer, Matthias Eiber, Matthew R. Cooperberg, Mehrdad Alemozzaffar, Matthew K. Tollefson, Scott E. Delacroix, and Brian F. Chapin. "The use of PET/CT in prostate cancer." Prostate cancer and prostatic diseases 21, no. 1 (2018): 4-21.
18. Rayn, Kareem N., Youssef A. Elnabawi, and Niki Sheth. "Clinical implications of PET/CT in prostate cancer management." Translational andrology and urology 7, no. 5 (2018): 844.
19. Reske, Sven N., Norbert M. Blumstein, and Gerhard Glatting. "[11 C] choline PET/CT imaging in occult local relapse of prostate cancer after radical prostatectomy." European journal of nuclear medicine and molecular imaging 35, no. 1 (2008): 9-17.
20. Schuster, David M., Peter T. Nieh, Ashesh B. Jani, Rianot Amzat, F. DuBois Bowman, Raghuveer K. Halkar, Viraj A. Master et al. "Anti-3-[18F] FACBC positron emission tomography-computerized tomography and 111In-capromab pendetide single photon emission computerized tomography-computerized tomography for recurrent prostate carcinoma: results of a prospective clinical trial." The Journal of urology 191, no. 5 (2014): 1446-1453.
21. Wondergem, Maurits, Friso M. van der Zant, Tjeerd van der Ploeg, and Remco JJ Knol. "A literature review of 18F-fluoride PET/CT and 18F-choline or 11C-choline PET/CT for detection of bone metastases in patients with prostate cancer." Nuclear medicine communications 34, no. 10 (2013): 935-945.
22. Nanni, Cristina, Lucia Zanoni, Cristian Pultrone, Riccardo Schiavina, Eugenio Brunocilla, Filippo Lodi, Claudio Malizia et al. "18 F-FACBC (anti1-amino-3-18 F-fluorocyclobutane-1-carboxylic acid) versus 11 C-choline PET/CT in prostate cancer relapse: results of a prospective trial." European journal of nuclear medicine and molecular imaging 43, no. 9 (2016): 1601-1610.
23. Calais, Jeremie, Francesco Ceci, Matthias Eiber, Thomas A. Hope, Michael S. Hofman, Christoph Rischpler, Tore Bach-Gansmo et al. "18F-fluciclovine PET-CT and 68Ga-PSMA-11 PET-CT in patients with early biochemical recurrence after prostatectomy: a prospective, single-centre, single-arm, comparative imaging trial." The Lancet Oncology 20, no. 9 (2019): 1286-1294.
24. Eiber, Matthias, Gregor Weirich, Konstantin Holzapfel, Michael Souvatzoglou, Bernhard Haller, Isabel Rauscher, Ambros J. Beer et al. "Simultaneous 68Ga-PSMA HBED-CC PET/MRI improves the localization of primary prostate cancer." European urology 70, no. 5 (2016): 829-836.
25. Zippel, Claus, Sarah C. Ronski, Sabine Bohnet-Joschko, Frederik L. Giesel, and Klaus Kopka. "Current Status of PSMA-Radiotracers for Prostate Cancer: Data Analysis of Prospective Trials Listed on ClinicalTrials. gov." Pharmaceuticals 13, no. 1 (2020): 12.
26. Klotz, Laurence, Greg Pond, Andrew Loblaw, Linda Sugar, Madeline Moussa, David Berman, Theo Van der Kwast et al. "Randomized Study of Systematic Biopsy Versus Magnetic Resonance Imaging and Targeted and Systematic Biopsy in Men on Active Surveillance (ASIST): 2-year Postbiopsy Follow-up." European urology (2019).

Localized Prostate Cancer Management in the Time of COVID-19

The rapid spread of Coronavirus Disease 2019 (COVID-19) throughout the world, caused by the betacoronavirus SARS-CoV-2, has had dramatic effects on health care systems with impacts far beyond the patients actually infected with COVID-19. Patients who manifest severe forms of COVID-19 requiring respiratory support typically require this for prolonged durations, with a mean of 13 days of respiratory support reported by the China Medical Treatment Expert Group for COVID-19.1 This lengthy requirement for ventilator support and ICU resources, exacerbated by relatively little excess health system capacity to accommodate epidemics, means that health care systems can (and have in the case of many hospitals in Italy) become overwhelmed relatively quickly.
Written by: Zachary Klaassen, MD, MSc
References: 1. Guan, Wei-jie, Zheng-yi Ni, Yu Hu, Wen-hua Liang, Chun-quan Ou, Jian-xing He, Lei Liu et al. "Clinical characteristics of coronavirus disease 2019 in China." New England Journal of Medicine (2020).
2. March 13, Online, and 2020. “COVID-19: Recommendations for Management of Elective Surgical Procedures.” American College of Surgeons. Accessed April 10, 2020. https://www.facs.org/covid-19/clinical-guidance/elective-surgery.
3. March 17, Online, and 2020. “COVID-19: Guidance for Triage of Non-Emergent Surgical Procedures.” American College of Surgeons. Accessed April 10, 2020. https://www.facs.org/covid-19/clinical-guidance/triage.
4. Liang, Wenhua, Weijie Guan, Ruchong Chen, Wei Wang, Jianfu Li, Ke Xu, Caichen Li et al. "Cancer patients in SARS-CoV-2 infection: a nationwide analysis in China." The Lancet Oncology 21, no. 3 (2020): 335-337.
5. Choo, Richard, Laurence Klotz, Cyril Danjoux, Gerard C. Morton, Gerrit DeBoer, Ewa Szumacher, Neil Fleshner, Peter Bunting, and George Hruby. "Feasibility study: watchful waiting for localized low to intermediate grade prostate carcinoma with selective delayed intervention based on prostate specific antigen, histological and/or clinical progression." The Journal of urology 167, no. 4 (2002): 1664-1669.
6. Klotz, Laurence, Danny Vesprini, Perakaa Sethukavalan, Vibhuti Jethava, Liying Zhang, Suneil Jain, Toshihiro Yamamoto, Alexandre Mamedov, and Andrew Loblaw. "Long-term follow-up of a large active surveillance cohort of patients with prostate cancer." Journal of Clinical Oncology 33, no. 3 (2015): 272-277.
7. Musunuru, Hima Bindu, Toshihiro Yamamoto, Laurence Klotz, Gabriella Ghanem, Alexandre Mamedov, Peraka Sethukavalan, Vibhuti Jethava et al. "Active surveillance for intermediate risk prostate cancer: survival outcomes in the Sunnybrook experience." The Journal of urology 196, no. 6 (2016): 1651-1658.
8. Wilt, Timothy J., Tien N. Vo, Lisa Langsetmo, Philipp Dahm, Thomas Wheeler, William J. Aronson, Matthew R. Cooperberg, Brent C. Taylor, and Michael K. Brawer. "Radical Prostatectomy or Observation for Clinically Localized Prostate Cancer: Extended Follow-up of the Prostate Cancer Intervention Versus Observation Trial (PIVOT)." European urology (2020).
9. Bourgade, Vincent, Sarah J. Drouin, David R. Yates, Jerôme Parra, Marc-Olivier Bitker, Olivier Cussenot, and Morgan Rouprêt. "Impact of the length of time between diagnosis and surgical removal of urologic neoplasms on survival." World journal of urology 32, no. 2 (2014): 475-479.
10. Vickers, Andrew J., Fernando J. Bianco Jr, Stephen Boorjian, Peter T. Scardino, and James A. Eastham. "Does a delay between diagnosis and radical prostatectomy increase the risk of disease recurrence?." Cancer: Interdisciplinary International Journal of the American Cancer Society 106, no. 3 (2006): 56-580.
11. Korets, Ruslan, Catherine M. Seager, Max S. Pitman, Gregory W. Hruby, Mitchell C. Benson, and James M. McKiernan. "Effect of delaying surgery on radical prostatectomy outcomes: a contemporary analysis." BJU international 110, no. 2 (2012): 211-216.
12. van den Bergh, Roderick CN, Ewout W. Steyerberg, Ali Khatami, Gunnar Aus, Carl Gustaf Pihl, Tineke Wolters, Pim J. van Leeuwen, Monique J. Roobol, Fritz H. Schröder, and Jonas Hugosson. "Is delayed radical prostatectomy in men with low‐risk screen‐detected prostate cancer associated with a higher risk of unfavorable outcomes?." Cancer: Interdisciplinary International Journal of the American Cancer Society 116, no. 5 (2010): 1281-1290.
13. van den Bergh, Roderick CN, Peter C. Albertsen, Chris H. Bangma, Stephen J. Freedland, Markus Graefen, Andrew Vickers, and Henk G. van der Poel. "Timing of curative treatment for prostate cancer: a systematic review." European urology 64, no. 2 (2013): 204-215.
14. Cooperberg, Matthew R., and Peter R. Carroll. "Trends in management for patients with localized prostate cancer, 1990-2013." Jama 314, no. 1 (2015): 80-82.
15. Gupta, Natasha, Trinity J. Bivalacqua, Misop Han, Michael A. Gorin, Ben J. Challacombe, Alan W. Partin, and Mufaddal K. Mamawala. "Evaluating the impact of length of time from diagnosis to surgery in patients with unfavourable intermediate‐risk to very‐high‐risk clinically localised prostate cancer." BJU international 124, no. 2 (2019): 268-274.
16. Patel, Premal, Ryan Sun, Benjamin Shiff, Kiril Trpkov, and Geoffrey Thomas Gotto. "The effect of time from biopsy to radical prostatectomy on adverse pathologic outcomes." Research and reports in urology 11 (2019): 53.
17. Aas, Kirsti, Sophie Dorothea Fosså, Rune Kvåle, Bjørn Møller, Tor Åge Myklebust, Ljiljana Vlatkovic, Stig Müller, and Viktor Berge. "Is time from diagnosis to radical prostatectomy associated with oncological outcomes?." World journal of urology 37, no. 8 (2019): 1571-1580.
18. Fossati, Nicola, Martina Sofia Rossi, Vito Cucchiara, Giorgio Gandaglia, Paolo Dell’Oglio, Marco Moschini, Nazareno Suardi et al. "Evaluating the effect of time from prostate cancer diagnosis to radical prostatectomy on cancer control: can surgery be postponed safely?." In Urologic Oncology: Seminars and Original Investigations, vol. 35, no. 4, pp. 150-e9. Elsevier, 2017.
19. Berg, William T., Matthew R. Danzig, Jamie S. Pak, Ruslan Korets, Arindam RoyChoudhury, Gregory Hruby, Mitchell C. Benson, James M. McKiernan, and Ketan K. Badani. "Delay from biopsy to radical prostatectomy influences the rate of adverse pathologic outcomes." The Prostate 75, no. 10 (2015): 1085-1091.
20. Meunier, M. E., Y. Neuzillet, C. Radulescu, C. Cherbonnier, J. M. Hervé, M. Rouanne, V. Molinié, and T. Lebret. "Does the delay from prostate biopsy to radical prostatectomy influence the risk of biochemical recurrence?." Progres en urologie: journal de l'Association francaise d'urologie et de la Societe francaise d'urologie 28, no. 10 (2018): 475-481.
21. Zanaty, Marc, Mansour Alnazari, Kelsey Lawson, Mounsif Azizi, Emad Rajih, Abdullah Alenizi, Pierre-Alain Hueber et al. "Does surgical delay for radical prostatectomy affect patient pathological outcome? A retrospective analysis from a Canadian cohort." Canadian Urological Association Journal 11, no. 8 (2017): 265.
22. Zanaty, Marc, Mansour Alnazari, Khaled Ajib, Kelsey Lawson, Mounsif Azizi, Emad Rajih, Abdullah Alenizi et al. "Does surgical delay for radical prostatectomy affect biochemical recurrence? A retrospective analysis from a Canadian cohort." World journal of urology 36, no. 1 (2018): 1-6.
23. Westerman, Mary E., Vidit Sharma, George C. Bailey, Stephen A. Boorjian, Igor Frank, Matthew T. Gettman, R. Houston Thompson, Matthew K. Tollefson, and Robert Jeffrey Karnes. "Impact of time from biopsy to surgery on complications, functional and oncologic outcomes following radical prostatectomy." International braz j urol 45, no. 3 (2019): 468-477.
24. Martin, George L., Rafael N. Nunez, Mitchell D. Humphreys, Aaron D. Martin, Robert G. Ferrigni, Paul E. Andrews, and Erik P. Castle. "Interval from prostate biopsy to robot‐assisted radical prostatectomy: effects on perioperative outcomes." BJU international 104, no. 11 (2009): 1734-1737.
25. Schifano, N., P. Capogrosso, E. Pozzi, E. Ventimiglia, W. Cazzaniga, R. Matloob, G. Gandaglia et al. "Impact of time from diagnosis to treatment on erectile function outcomes after radical prostatectomy." Andrology 8, no. 2 (2020): 337-341.
26. Radomski, Lenny, Johan Gani, Greg Trottier, and Antonio Finelli. "Active surveillance failure for prostate cancer: does the delay in treatment increase the risk of urinary incontinence?." The Canadian journal of urology 19, no. 3 (2012): 6287-6292.
27. Kumar, Satish, Mike Shelley, Craig Harrison, Bernadette Coles, Timothy J. Wilt, and Malcolm Mason. "Neo‐adjuvant and adjuvant hormone therapy for localised and locally advanced prostate cancer." Cochrane Database of Systematic Reviews 4 (2006).

Androgen Receptor Signaling in Castration-Resistant Prostate Cancer

While androgen deprivation therapy (ADT) is nearly ubiquitously successful in suppressing testosterone to castrate levels with resultant declines in prostate-specific antigen (PSA) levels, the development of resistance is nearly as inevitable. While the natural history is variable, evidence suggests that most patients with advanced or metastatic prostate cancer will have disease progression within two to three years after initiation of androgen deprivation therapy, resulting in so-called “castration-resistant prostate cancer.”
Written by: Zachary Klaassen, MD, MSc
References: 1. Huggins, Charles, and Clarence V. Hodges. "Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate." The Journal of urology 167, no. 2 Part 2 (2002): 948-951.
2. Coutinho, Isabel, Tanya K. Day, Wayne D. Tilley, and Luke A. Selth. "Androgen receptor signaling in castration-resistant prostate cancer: a lesson in persistence." Endocrine-related cancer 23, no. 12 (2016): T179-T197.
3. Visakorpi, Tapio, Eija Hyytinen, Pasi Koivisto, Minna Tanner, Riitta Keinänen, Christian Palmberg, Aarno Palotie, Teuvo Tammela, Jorma Isola, and Olli-P. Kallioniemi. "In vivo amplification of the androgen receptor gene and progression of human prostate cancer." Nature genetics 9, no. 4 (1995): 401-406.
4. Chen, Charlie D., Derek S. Welsbie, Chris Tran, Sung Hee Baek, Randy Chen, Robert Vessella, Michael G. Rosenfeld, and Charles L. Sawyers. "Molecular determinants of resistance to antiandrogen therapy." Nature medicine 10, no. 1 (2004): 33-39.
5. Wyatt, Alexander W., and Martin E. Gleave. "Targeting the adaptive molecular landscape of castration‐resistant prostate cancer." EMBO molecular medicine 7, no. 7 (2015): 878-894.
6. Robinson, Dan, Eliezer M. Van Allen, Yi-Mi Wu, Nikolaus Schultz, Robert J. Lonigro, Juan-Miguel Mosquera, Bruce Montgomery et al. "Integrative clinical genomics of advanced prostate cancer." Cell 161, no. 5 (2015): 1215-1228.
7. Grasso, Catherine S., Yi-Mi Wu, Dan R. Robinson, Xuhong Cao, Saravana M. Dhanasekaran, Amjad P. Khan, Michael J. Quist et al. "The mutational landscape of lethal castration-resistant prostate cancer." Nature 487, no. 7406 (2012): 239-243.
8. Cai, Changmeng, Housheng Hansen He, Sen Chen, Ilsa Coleman, Hongyun Wang, Zi Fang, Shaoyong Chen et al. "Androgen receptor gene expression in prostate cancer is directly suppressed by the androgen receptor through recruitment of lysine-specific demethylase 1." Cancer cell 20, no. 4 (2011): 457-471.
9. Antonarakis, Emmanuel S., Changxue Lu, Hao Wang, Brandon Luber, Mary Nakazawa, Jeffrey C. Roeser, Yan Chen et al. "AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer." New England Journal of Medicine 371, no. 11 (2014): 1028-1038.
10. Chmelar, Renée, Grant Buchanan, Eleanor F. Need, Wayne Tilley, and Norman M. Greenberg. "Androgen receptor coregulators and their involvement in the development and progression of prostate cancer." International Journal of cancer 120, no. 4 (2007): 719-733.
11. Zoubeidi, Amina, Anousheh Zardan, Eliana Beraldi, Ladan Fazli, Richard Sowery, Paul Rennie, Colleen Nelson, and Martin Gleave. "Cooperative interactions between androgen receptor (AR) and heat-shock protein 27 facilitate AR transcriptional activity." Cancer research 67, no. 21 (2007): 10455-10465.
12. Gregory, Christopher W., Bin He, Raymond T. Johnson, O. Harris Ford, James L. Mohler, Frank S. French, and Elizabeth M. Wilson. "A mechanism for androgen receptor-mediated prostate cancer recurrence after androgen deprivation therapy." Cancer research 61, no. 11 (2001): 4315-4319.
13. Qin, Jun, Hui-Ju Lee, San-Pin Wu, Shih-Chieh Lin, Rainer B. Lanz, Chad J. Creighton, Francesco J. DeMayo, Sophia Y. Tsai, and Ming-Jer Tsai. "Androgen deprivation–induced NCoA2 promotes metastatic and castration-resistant prostate cancer." The Journal of clinical investigation 124, no. 11 (2014): 5013-5026.

Complications & Adverse Events – External Urinary Catheters

An external urine collection device (EUCD) may be external and less invasive but they are not free of risks. Complications and adverse effects include skin lesion/ulceration and breakdown from pressure necrosis and moisture, urethral fistula or very rarely, gangrene of the penis. The majority of complications involve perineal/genital skin issues, primarily occurring in 15-30% of male patients and involve external penile shaft problems.

Written by: Diane K. Newman, DNP, ANP-BC, FAAN
References: 1. Al-Awadhi, N. M., N. Al-Brahim, M. S. Ahmad, and E. Yordanov. "Giant fibroepithelial polyp of the penis associated with long-term use of condom catheter. Case report and literature review." The Canadian journal of urology 14, no. 4 (2007): 3656-3659.
2. Banerji, John S., Sanjeev Shah, and Nitin S. Kekre. "Fibroepithelial polyp of the prepuce: A rare complication of long-term condom catheter usage." Indian journal of urology: IJU: journal of the Urological Society of India 24, no. 2 (2008): 263.
3. Beeson, Terrie, and Carmen Davis. "Urinary management with an external female collection device." Journal of Wound, Ostomy, and Continence Nursing 45, no. 2 (2018): 187.
4. Bycroft, J., R. Hamid, and P. J. R. Shah. "Penile erosion in spinal cord injury–an important lesson." Spinal cord 41, no. 11 (2003): 643-644.
5. Golji, Hossein. "Complications of external condom drainage." Paraplegia 19, no. 3 (1981): 189-197.
6. Grigoryan, Larissa, Michael S. Abers, Quratulain F. Kizilbash, Nancy J. Petersen, and Barbara W. Trautner. "A comparison of the microbiologic profile of indwelling versus external urinary catheters." American journal of infection control 42, no. 6 (2014): 682-684.
7. Harmon, Christopher B., Suzanne M. Connolly, and Thayne R. Larson. "Condom-related allergic contact dermatitis." The Journal of urology 153, no. 4 (1995): 1227-1228.
8. Newman, D. K. "Devices, products, catheters, and catheter-associated urinary tract infections." Core curriculum for urologic nursing. 1st ed. Pitman: Society of Urologic Nurses and Associates, Inc (2017): 439-66.
9. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.
10. Milanesi, Nicola, Gastone Bianchini, Angelo Massimiliano D'ERME, and Stefano Francalanci. "Allergic reaction to condom catheter for bladder incontinence." Contact dermatitis 69, no. 3 (2013): 182-183.

First Line Therapy for Metastatic Clear Cell Renal Cell Carcinoma

As previous UroToday Center of Excellence articles have highlighted, clear cell renal cell carcinoma (ccRCC) is the most common histologic subtype of renal cell carcinoma (RCC). Likely due to its much higher prevalence, the vast majority of systemic therapies for RCC have been investigated among patients with ccRCC.

Second perhaps only to advanced prostate cancer, the metastatic clear cell renal cell carcinoma disease space has undergone rapid and transformational change over the past fifteen years. This rapidly shifting treatment landscape was highlighted recently at the American Society of Clinical Oncology 2019 Annual Meeting:

figure-1-treatment-landscape-metastatic-RCC2x.jpg

Early years: cytokine therapy


It has been recognized for many decades that renal cell carcinoma is an immunologically active tumor. As a result, modulators of the immune system were among the first therapeutic targets for advanced ccRCC. Prior to 2005, treatment for metastatic RCC (mRCC) was limited to cytokine therapies (interferon-alfa and interleukin-2).

Interferon-α was one of the first cytokines assessed for the treatment of metastatic ccRCC. Based on early data suggesting a response rate between 10 to 15%5 and comparative data demonstrating a survival benefit compared to other available systemic therapies available at the time,6 interferon-alfa retained utilization despite significant toxicity. Further, it was among patients with metastatic RCC receiving interferon-alfa that the Motzer prognostic criteria were derived.6 In their seminal paper in the Journal of Clinical Oncology, Motzer and colleagues demonstrated that low Karnofsky performance status, high lactate dehydrogenase, low serum hemoglobin, high corrected serum calcium, and short time from initial RCC diagnosis to start of interferon-alfa therapy (<1 year) could be used to risk-stratify patients with renal cell carcinoma. However, even among patients treated at a center of excellence, median overall survival was only 30 months in favorable-risk patients, 14 months in intermediate-risk patients and five months in poor-risk patients.6

Other immunologic therapies were explored including interleukin-2. While response rates were similar to interferon-based therapies (~15 to 20%),7 interleukin-2 was distinct in that durable complete responses were observed in approximately 7 to 9% of patients.8 This observation led to the U.S. Food and Drug Administration (FDA) approval of high-dose IL-2 in 1992. However, IL-2 is associated with significant toxicity which has limited its widespread use.

Combinations of interferon and interleukin therapies were explored subsequently those these data demonstrated no improvement in overall survival,9 with significantly increased toxicity compared to monotherapy with either agent.

A new standard: molecularly targeted agents


Based on work into the molecular biology underlying ccRCC, researchers were led to “rational targeted therapeutics” including targeting of the vascular endothelial growth factor (VEGF) pathway and mammalian target of rapamycin (mTOR). Mammalian target of rapamycin (mTOR) plays a key role in regulating HIF-α, thus modulating the pathway between abnormalities in VHF and proliferation.

Bevacizumab, a humanized monoclonal antibody against VEGF-A, was the first inhibitor of the VEGF pathway used in clinical trials. As is standard in an oncology pathway, it was first tested in patients who had progressed on the current standard of care (cytokine therapy) and subsequently tested in untreated patients. In head-to-head trials against interferon-alfa, the addition of bevacizumab to interferon resulted in significant improvements in response rate and progression-free survival.10,11 Today, bevacizumab is uncommonly used as monotherapy in untreated patients but is considered as second-line therapy in patients who have failed prior therapy with tyrosine kinase inhibitors.

Tyrosine-kinase inhibitors also target the VEGF pathway, through inhibition of a combination of VEGFR-2, PDGFR-β, raf-1 c-Kit, and Flt3 (sunitinib and sorafenib). In 2006, sorafenib was shown to have biologic activity in ccRCC. Subsequent studies demonstrated improvements in progression-free survival compared with placebo in patients who have previously failed cytokine therapy and improvements in tumor regression compared to interferon in previously untreated patients. As highlighted in the Figure above, sorafenib was one the first molecularly targeted agents clinically available. However, despite FDA approval, sorafenib was quickly supplanted by sunitinib as a first-line VEGF inhibitor.

In keeping with the aforementioned oncology pipeline, sunitinib was first evaluated among patients who had previously received cytokine treatment. Subsequently, it was compared to interferon-α in a pivotal Phase III randomized trial.12 Among 750 patients with previously untreated, metastatic RCC randomized, median progression-free survival was significantly longer among those who received sunitinib (11 months) than those who received interferon-alfa (5 months; hazard ratio 0.42, 95% confidence interval 0.32 to 0.54). Similar benefits were seen in the overall response rate with subsequent follow-up demonstrating a strong trend towards improved overall survival. In the pivotal trial, patients who received sunitinib had a significantly better quality of life than those who received interferon-alfa12, despite class-based toxicity profile including gastrointestinal events, dermatologic complications including hand-foot desquamation, hypertension, and general malaise. On account of these data, sunitinib is widely used as a first-line treatment of RCC.

Since the approval of sunitinib and sorafenib, there has been the development and subsequent approval of a number of other tyrosine kinase inhibitors. For the most part, the goal of these agents has been to reduce the toxicity of VEGF inhibitors while retaining oncologic efficacy. Comparative data of pazopanib and sunitinib have demonstrated non-inferior oncologic outcomes with decreased toxicity among patients receiving pazopanib.13 Axitinib was evaluated first as second-line therapy14 and then in the first-line setting compared to sorafenib.15 Among 192 patients with previously untreated ccRCC randomized to axitinib and 96 patients randomized to sorafenib, median progression-free survival was not significantly different (10.1 months and 6.5 months, respectively; hazard ratio 0.77, 95% confidence interval 0.56 to 1.05)15. Finally, tivozanib has been compared to sorafenib among patients who had not previously received VEGF or mTOR-targeting therapies. While this study demonstrated tivozanib’s activity, it was not FDA approved and is therefore not used.

Most recently, a multikinase inhibitor, cabozantinib has been approved for the first-line treatment of mRCC. In the Phase II CABOSUN trial, cabozantinib was compared to sunitinib in the first-line treatment of patients with intermediate or poor-risk mRCC.16 Assessing the primary outcome of progression-free survival, the 79 patients randomized to cabozantinib had significantly longer progression-free survival (8.2 months) compared to the 78 randomized to sunitinib (5.6 months; hazard ratio 0.66, 95% confidence interval 0.46 to 0.95). A recent update on this trial utilizing independent progression-free survival (PFS) review demonstrated comparable results (hazard ratio 0.48, 95% confidence interval 0.31 to 0.74)17. Even with an increased follow-up (median 34.5 months), no significant difference in overall survival was demonstrated (26.6 months in patients receiving cabozantinib and 21.2 months in those receiving sunitinib; hazard ratio 0.80, 95% confidence interval 0.53 to 1.21). While this appears to demonstrate a significant benefit to cabozantinib, median survival in the sunitinib arm was lower than may be expected18 which would serve to exaggerate the apparent benefit of cabozantinib.

In parallel to the development, clinical appraisal and utilization of VEGF inhibitors have come the development of mTOR inhibitors. Temsirolimus was the first mTOR inhibitor to reach clinical utility in patients with metastatic RCC. In the Global ARCC Trial, temsirolimus, interferon, and the combination were compared among 626 patients with pre-defined poor-risk metastatic RCC who had not previously received systemic therapy.19 Notable compared to many trials in this disease space that have utilized progression-free survival as the primary outcome, overall survival was the primary outcome, with the study powered based on comparisons of the temsirolimus group and the combination group to the interferon-alfa group. Patients who received temsirolimus had significantly improved overall survival compared to those receiving interferon-alfa (hazard ratio 0.73, 95% confidence interval 0.58 to 0.92). Notably, the combination arm did not offer a benefit compared to interferon alone. Unlike temsirolimus which must be administered intravenously, everolimus is an oral agent.

What’s old is new: immunotherapy for RCC


The immunologic basis for the treatment of advanced RCC has been well established, including the aforementioned cytokine therapies. Thus, it should not be surprising that the use of checkpoint inhibitors has demonstrated benefit in patients with metastatic RCC.

First presented at ESMO in the fall of 2017 and subsequently published in the spring of 2018, CheckMate 214 demonstrated an overall survival (OS) benefit for first-line nivolumab plus ipilimumab vs sunitinib.20 This trial randomized 1096 patients to the combination immunotherapy approach of nivolumab plus ipilimumab (550 patients) or sunitinib (546 patients). The majority of patients had intermediate or poor-risk disease (n=847). Overall survival was significantly improved in the overall patient population; however, stratified analyses provide more nuanced results. Among the subgroup of patients with intermediate or poor-risk RCC, treatment with nivolumab plus ipilimumab resulted in significantly improved overall response rate, comparable progression-free survival, and significantly improved overall survival. In contrast, among patients with favorable-risk disease, progression-free survival and overall response rate were higher among patients who received sunitinib. Recently, Escudier and colleagues have assessed the efficacy of nivolumab and ipilimumab according to the number of IMDC risk factors.21 In keeping with the previously reported differences in the comparative benefit of nivo/ipi versus sunitinib on the basis of risk category (intermediate/poor versus favorable), the authors demonstrated stable overall response rate (ORR) across increasing numbers of IMDC risk factors (from zero to six) for those who received nivolumab and ipilimumab, while the ORR in patients treated with sunitinib decreased with an increasing number of IMDC risk factors.

The next frontier: combinations of targeted therapy and immunotherapy


Shortly after the data from CheckMate214 emerged, the results of IMmotion151 were presented at GU ASCO in the spring of 2018 and subsequently published. This Phase III trial compared first-line atezolizumab + bevacizumab versus sunitinib among 915 patients with previously untreated metastatic RCC.22 This regime was active with a significant benefit in progression-free survival (11.2 months versus 7.7 months; hazard ratio 0.74, 95% confidence interval 0.57 to 0.96) among the whole cohort of patients and had lower rates of significant (grade 3-4) adverse events (40% vs 54%). 

Since the publication of CheckMate214 and IMmotion151, two trials have reported on combinations of checkpoint inhibitors and tyrosine kinase inhibitors: KEYNOTE-426 and JAVELIN Renal 101.

In KEYNOTE-426, 861 patients with metastatic clear cell RCC who had not previously received systemic therapy were randomized to pembrolizumab plus axitinib or sunitinib and followed for the co-primary endpoints of overall survival and progression-free survival.23 Similar to CheckMate214, the majority of patients had intermediate or poor-risk disease. While median survival was not reached, patients who received pembrolizumab and axitinib had improved overall survival (hazard ratio 0.53, 95% confidence interval 0.38 to 0.74) and progression-free survival (hazard ratio 0.69, 95% confidence interval 0.57 to 0.84), as well as overall response rate. These results were consistent across subgroups of demographic characteristics, IMDC risk categories, and programmed death-ligand 1 (PD-L1) expression level. Grade 3 to 5 adverse events were somewhat more common among patients getting pembrolizumab and axitinib, though rates of discontinuation were lower.


Similarly, JAVELIN Renal 101 randomized 886 patients to avelumab and axitinib or sunitinib.24 Again, the preponderance of patients had IMDC intermediate or poor-risk disease. This analysis of the primary endpoints was progression-free survival and overall survival in patients with PD-L1 positive tumors. Notably, 560 of the 886 patients had PD-L1 positive tumors. Among the PD-L1 positive subgroup, progression-free survival (hazard ratio 0.61, 95% confidence interval 0.47 to 0.79) was improved in patients receiving avelumab and axitinib compared to sunitinib while overall survival did not significantly differ (hazard ratio 0.82, 95% confidence interval 0.53 to 1.28). In the overall study population, progression-free survival was similarly improved, as compared to the PD-L1 positive population (hazard ratio 0.69, 95% confidence interval 0.56 to 0.84).

Dead-ends


Numerous chemotherapeutic agents have been explored in ccRCC. These include 5-FU, gemcitabine, vinblastine, bleomycin, and platinum. Meta-analyses of these data demonstrate poor response25 and thus cytotoxic chemotherapy is not indicated in the treatment of advanced RCC. Similarly, hormonal therapies including medroxyprogesterone have been explored but have no role in the modern management of advanced RCC.

Integration of treatment options for patients with metastatic ccRCC


Due to the rapid proliferation of treatment options in first-line treatment of metastatic clear cell renal cell carcinoma, there is a paucity of direct comparative data. The majority of new agents have been compared to sunitinib which was the standard of care at the time that trials were designed. Due to the lack of comparative data, it may be difficult to ascertain which treatment to offer patients who present in clinic. There are a number of ways to approach this issue. First, one may take a quantitative approach, utilizing the available comparative data in a network meta-analysis; second, one may rely upon eminence, as in expert-informed guidelines; finally, one may rely on individual clinical experience.

Assessing this quantitatively, we have recently performed a network meta-analysis of first-line agents in metastatic RCC.26 Assessing agents which are commonly utilized in 2019, we examined 12 relevant trials. Depending on the outcome of interest (progression-free survival, overall survival, or adverse events), the preferred treatment varied. However, pembrolizumab and axitinib appeared to have a high likelihood of being preferred for oncologic outcomes.
Second, considering a panel of expert opinion, the European Association of Urology recently updated its guidelines on the treatment of renal cell carcinoma. Their recommendations are highlighted in the following figure, taken from the EAU guideline:

figure-2-EAU-guidelines-RCC-treatment2x.jpg
Notably, the most recent version of these guidelines alludes to the recently published data but have not yet integrated the role of atezolizumab plus bevacizumab, pembrolizumab plus axitinib, or avelumab plus axitinib in guideline recommendations.

Finally, we may rely on the guidance of individual clinical experience.

What about surgery?


The role of cytoreductive nephrectomy in the management of metastatic renal cell carcinoma has dramatically changed with the publication of the CARMENA and SURTIME studies. The available evidence suggests that systemic therapy should be prioritized ahead of cytoreductive nephrectomy. However, there remains a role of cytoreductive nephrectomy in select patients.

Published Date: March 17th, 2020

Written by: Zachary Klaassen, MD, MSc
References: 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: a cancer journal for clinicians. 2018;68(1):7-30.
2. Welch HG, Skinner JS, Schroeck FR, Zhou W, Black WC. Regional Variation of Computed Tomographic Imaging in the United States and the Risk of Nephrectomy. JAMA internal medicine. 2018;178(2):221-227.
3. Motzer RJ, Mazumdar M, Bacik J, Berg W, Amsterdam A, Ferrara J. Survival and prognostic stratification of 670 patients with advanced renal cell carcinoma. Journal of Clinical Oncology. 1999;17:2530-2540.
4. Negrier S, Escudier B, Gomez F, et al. Prognostic factors of survival and rapid progression in 782 patients with metastatic renal carcinomas treated by cytokines: a report from the Groupe Francais d'Immunotherapie. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2002;13(9):1460-1468.
5. Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2002;20(1):289-296.
6. Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev. 2005(1):CD001425.
7. Dutcher JP, Atkins M, Fisher R, et al. Interleukin-2-based therapy for metastatic renal cell cancer: the Cytokine Working Group experience, 1989-1997. Cancer J Sci Am. 1997;3 Suppl 1:S73-78.
8. Rosenberg SA, Yang JC, White DE, Steinberg SM. Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2: identification of the antigens mediating response. Ann Surg. 1998;228(3):307-319.
9. Negrier S, Escudier B, Lasset C, et al. Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal-cell carcinoma. Groupe Francais d'Immunotherapie. The New England journal of medicine. 1998;338(18):1272-1278.
10. Rini BI, Halabi S, Rosenberg JE, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2008;26(33):5422-5428.
11. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370(9605):2103-2111.
12. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. The New England journal of medicine. 2007;356(2):115-124.
13. Motzer RJ, Hutson TE, Cella D, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. The New England journal of medicine. 2013;369(8):722-731.
14. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011;378(9807):1931-1939.
15. Hutson TE, Lesovoy V, Al-Shukri S, et al. Axitinib versus sorafenib as first-line therapy in patients with metastatic renal-cell carcinoma: a randomised open-label phase 3 trial. The lancet oncology. 2013;14(13):1287-1294.
16. Choueiri TK, Halabi S, Sanford BL, et al. Cabozantinib Versus Sunitinib As Initial Targeted Therapy for Patients With Metastatic Renal Cell Carcinoma of Poor or Intermediate Risk: The Alliance A031203 CABOSUN Trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(6):591-597.
17. Choueiri TK, Hessel C, Halabi S, et al. Cabozantinib versus sunitinib as initial therapy for metastatic renal cell carcinoma of intermediate or poor risk (Alliance A031203 CABOSUN randomised trial): Progression-free survival by independent review and overall survival update. European journal of cancer. 2018;94:115-125.
18. Buti S, Bersanelli M. Is Cabozantinib Really Better Than Sunitinib As First-Line Treatment of Metastatic Renal Cell Carcinoma? Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2017;35(16):1858-1859.
19. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. The New England journal of medicine. 2007;356(22):2271-2281.
20. Escudier B, Tannir NM, McDermott D, et al. LBA5 - CheckMate 214: Efficacy and safety of nivolumab 1 ipilimumab (N1I) v sunitinib (S) for treatment-naive advanced or metastatic renal cell carcinoma (mRCC), including IMDC risk and PD-L1 expression subgroups. Annals of Oncology. 2017;28(Supplement 5):621-622.
21. Escudier B, Motzer RJ, Tannir NM, et al. Efficacy of Nivolumab plus Ipilimumab According to Number of IMDC Risk Factors in CheckMate 214. European urology. 2019.
22. Motzer R, Powles T, Atkins M, et al. IMmotion151: A Randomized Phase III Study of Atezolizumab Plus Bevacizumab vs Sunitinib in Untreated Metastatic Renal Cell Carcinoma. Journal of Clinical Oncology. 2018;36(Suppl 6S).
23. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. The New England journal of medicine. 2019;380(12):1116-1127.
24. Motzer RJ, Penkov K, Haanen J, et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. The New England journal of medicine. 2019;380(12):1103-1115.
25. Yagoda A, Abi-Rached B, Petrylak D. Chemotherapy for advanced renal-cell carcinoma: 1983-1993. Semin Oncol. 1995;22(1):42-60.
26. Hahn AW, Klaassen Z, Agarwal N, et al. First-line Treatment of Metastatic Renal Cell Carcinoma: A Systematic Review and Network Meta-analysis. Eur Urol Oncol. 2019;2(6):708-715.

Nephron-Sparing Approaches in Upper Tract Urothelial Carcinoma

Upper tract urothelial carcinoma, which may affect the renal pelvis or ureter, is a relatively rare disease, accounting for less than 10% of all urothelial carcinomas.1 The etiology of this uncommon cancer is discussed in more detail in a previous UroToday Center of Excellence article.

Written by: Zachary Klaassen, MD, MSc
References: 1. Siegel, Rebecca L., Kimberly D. Miller, and Ahmedin Jemal. "Cancer statistics, 2019." CA: a cancer journal for clinicians 69, no. 1 (2019): 7-34.
2. Vashistha, Vishal, Ahmad Shabsigh, and Debra L. Zynger. "Utility and diagnostic accuracy of ureteroscopic biopsy in upper tract urothelial carcinoma." Archives of pathology & laboratory medicine 137, no. 3 (2013): 400-407.
3. Honda, Yukiko, Yuko Nakamura, Jun Teishima, Keisuke Goto, Toru Higaki, Keigo Narita, Motonori Akagi et al. "Clinical staging of upper urinary tract urothelial carcinoma for T staging: Review and pictorial essay." International Journal of Urology 26, no. 11 (2019): 1024-1032.
4. Rouprêt, Morgan, Marko Babjuk, Eva Compérat, Richard Zigeuner, Richard J. Sylvester, Maximilian Burger, Nigel C. Cowan et al. "European association of urology guidelines on upper urinary tract urothelial carcinoma: 2017 update." European urology 73, no. 1 (2018): 111-122.
5. Petros, Firas G., Roger Li, and Surena F. Matin. "Endoscopic approaches to upper tract urothelial carcinoma." Urologic Clinics 45, no. 2 (2018): 267-286.
6. Samson, Patrick, Arthur D. Smith, David Hoenig, and Zeph Okeke. "Endoscopic Management of Upper Urinary Tract Urothelial Carcinoma." Journal of endourology 32, no. S1 (2018): S-10.
7. Cutress, Mark L., Grant D. Stewart, Paimaun Zakikhani, Simon Phipps, Ben G. Thomas, and David A. Tolley. "Ureteroscopic and percutaneous management of upper tract urothelial carcinoma (UTUC): systematic review." BJU international 110, no. 5 (2012): 614-628.
8. Babjuk, Marko, Maximilian Burger, Eva M. Compérat, Paolo Gontero, A. Hugh Mostafid, Joan Palou, Bas WG van Rhijn et al. "European Association of Urology guidelines on non-muscle-invasive bladder cancer (TaT1 and carcinoma In Situ)-2019 update." European urology (2019).
9. Rastinehad, Ardeshir R., and Arthur D. Smith. "Bacillus Calmette-Guerin for upper tract urothelial cancer: is there a role?." Journal of endourology 23, no. 4 (2009): 563-568.
10. Nepple, Kenneth G., Fadi N. Joudi, and Michael A. O'Donnell. "Review of topical treatment of upper tract urothelial carcinoma." Advances in urology 2009 (2009).
11. Donin, Nicholas M., Sandra Duarte, Andrew T. Lenis, Randy Caliliw, Cristobal Torres, Anthony Smithson, Dalit Strauss-Ayali et al. "Sustained-release formulation of mitomycin C to the upper urinary tract using a thermosensitive polymer: a preclinical study." Urology 99 (2017): 270-277.
12. Williams, Steve K., Karin J. Denton, Andrea Minervini, Jon Oxley, Jay Khastigir, Anthony G. Timoney, and Francis X. Keeley. "Correlation of upper-tract cytology, retrograde pyelography, ureteroscopic appearance, and ureteroscopic biopsy with histologic examination of upper-tract transitional cell carcinoma." Journal of endourology 22, no. 1 (2008): 71-76.
13. de Bruijn, Ernst A., Harm P. Sleeboom, Peter JRO van Helsdingen, Allan T. van Oosterom, Ubbo R. Tjaden, and Robert AA Maes. "Pharmacodynamics and pharmacokinetics of intravesical mitomycin C upon different dwelling times." International journal of cancer 51, no. 3 (1992): 359-364.
14. Donin, Nicholas M., Dalit Strauss-Ayali, Yael Agmon-Gerstein, Nadav Malchi, Andrew T. Lenis, Stuart Holden, Allan J. Pantuck, Arie S. Belldegrun, and Karim Chamie. "Serial retrograde instillations of sustained release formulation of mitomycin C to the upper urinary tract of the Yorkshire swine using a thermosensitive polymer: safety and feasibility." In Urologic Oncology: Seminars and Original Investigations, vol. 35, no. 5, pp. 272-278. Elsevier, 2017.

Prevention of Skeletal-Related Events in Advanced Prostate Cancer

Patients with advanced prostate cancer are at significant risk of skeletal-related events (SREs) due to a complex interplay between bone health and prostate cancer due to cancer biology and the predilection of prostate cancer to spread to bone, the toxicity of prostate cancer treatments, and shared epidemiology of the two conditions.

Skeletal-related events are used to denote events related to osseous metastases, including pathologic bone fractures, spinal cord compression, orthopedic surgical intervention, and palliative radiation directed at the bone.1 This definition has been widely used in the design of randomized controlled trials and for drug approval. In some circumstances, authors include a change in systemic anti-neoplastic therapy as a result of bony pain in the definition of skeletal-related events. The key to this definition is that skeletal-related events may be clinically manifested due to symptoms or only radiographically detected. Thus, skeletal-related events may or may not be symptomatic. In contrast, symptomatic skeletal-related events (SSEs) is a relatively newer outcome representing a subset of skeletal-related events which symptomatically affect the patient experience. This outcome was first used in the Alpharadin in Symptomatic Prostate Cancer (ALSYMPCA) trial,2 wherein symptomatic skeletal-related events were defined as bone-directed radiotherapy to relieve bony pain, new symptomatic pathologic fractures, spinal cord compression, or tumor-related orthopedic surgery. While there is a significant overlap between these conditions, there are important differences that relate to both study design and patient care: namely, detection of SREs requires routine radiographic evaluation to detect asymptomatic skeletal-related events while detection of SSEs can be driven by patient evaluation.

Skeletal related events contribute significantly to the disease-related morbidity and mortality of prostate cancer, in addition to being very costly. A range of lifestyle, nutritional, and pharmaceutical interventions can be undertaken to decrease the risk of skeletal-related events in patients with advanced prostate cancer.

Lifestyle and nutrition-based interventions

These recommendations are not unique to patients with prostate cancer, but rather apply to all men with osteoporosis or decreased bone mineral density to reduce the risk of fractures.

The Endocrine Society Clinical Practice Guideline on Osteoporosis in Men recommends the following lifestyle interventions:3

  1. Calcium intake: it is recommended that men with or at risk for osteoporosis consume 1000 to 1200 mg of calcium daily. While dietary sources are preferable, calcium supplementation should be used if dietary calcium intake is inadequate.
  2. Vitamin D intake: men with low vitamin D levels (< 30 ng/mL or < 75 nmol/L) should receive vitamin D supplementation to raise serum vitamin D levels to at least these levels.
  3. Exercise: men at risk of osteoporosis are recommended to participate in weight-bearing activities at least three or four times per week, for 30 to 40 minutes per session.
  4. Alcohol intake: it is recommended that men at risk of osteoporosis reduce their alcohol intake to fewer than three units of alcohol. One unit of alcohol is defined as 10 mL of pure alcohol. This amount could be found in 25 mL of spirits (40% alcohol by volume), one third to one half a pint of beer (5-6% alcohol by volume), or half a standard glass of wine (12% alcohol by volume).
  5. Smoking cessation: it is recommended that all men at risk of osteoporosis stop smoking.

The American Urological Association 2018 Amendment of the Castration-Resistant Prostate Cancer similarly endorses preventative therapy in the form of supplemental calcium and vitamin D in men with castration-resistant prostate cancer.4 However, such interventions are likely advisable much earlier in the disease trajectory given the relatively minor risks of these supplements in contrast to the potentially debilitating and costly consequences of skeletal-related events. To this end, the NCCN and the National Osteoporosis Foundation recommend these interventions in all men over the age of 50 years who are receiving androgen deprivation therapy.

Pharmacologic interventions

In addition to calcium and vitamin D as highlighted above, the American Urological Association 2018 Amendment of the Castration-Resistant Prostate Cancer endorses pharmacotherapy with denosumab or zoledronic acid in men with castration-resistant prostate cancer.4

While further indications exist, the Endocrine Society Clinical Practice Guideline on Osteoporosis in Men recommends pharmacologic therapy in men at high risk for fracture based on, but not limited to, the following factors:3

  1. Men with a history of previous non-traumatic hip or vertebral fracture.
  2. Men with bone mineral density (T-score) that is 2.5 standard deviations or more below the mean of normal young white men, in the absence of spine or hip fractures.
  3. Men with a bone mineral density (T-score) from 1.0 to 2.5 standard deviations who also have an increased risk of fracture. In the United States, the suggested criteria for increased risk of fracture are a 10-year risk of any fracture equal to or exceeding 20% or a 10-year risk of hip fracture equal to or exceeding 3%, as determined using the FRAX tool. In other regions, the Guidelines recommend the utilization of region-specific guidelines.
  4. Men who are receiving long-term glucocorticoid treatment in significant doses defined by the 2010 American Society of Rheumatology as > 7.5 ng per day of prednisone or equivalent.

Pharmaceuticals - bisphosphonates

Bisphosphonates were among the first agents successfully used to prevent skeletal-related events in advanced prostate cancer. They function by reducing bone resorption through a variety of mechanisms including decreased osteoclast differentiation and survival and increased osteoblast survival. To do so, they compete with pyrophosphate for hydroxyapatite crystal binding sites, thus reducing osteoclast adherence to the bone. While there are a number of bisphosphonates used for a variety of clinical indications, zoledronic acid is most commonly used in patients with metastatic prostate cancer due to the Zometa 039 trial which demonstrated a reduction in SREs for patients who received zoledronic acid as compared to placebo.5 Based on these data, zoledronic acid is the only bisphosphonate approved for the prevention of skeletal-related events in men with metastatic prostate cancer.

Pharmaceuticals - denosumab

Denosumab is another agent which targets osteoclast activity. Bone exists in homeostasis between formation (driven by osteoblasts) and resorption (driven by osteoclasts). This homeostasis is regulated by the RANK (receptor activator of nuclear factor κB)/RANK-ligand system. Denosumab is a fully human monoclonal antibody that targets the RANK-ligand by mimicking OPG. Denosumab was first examined in postmenopausal women to prevent the development of osteoporosis and reduce the risk of fracture. Subsequently, denosumab was examined in women with breast cancer. Most relevantly, the Denosumab Protocol 20050103 compared denosumab and zoledronic acid in 1901 men with castrate-resistant prostate cancer.6 After a median follow-up of approximately one year, patients receiving denosumab had a significantly prolonged time to first-SRE (3.6 months incremental benefit, hazard ratio 0.82, 95% confidence interval 0.71 to 0.95). As a result of these data, denosumab was approved for the prevention of SREs in patients with metastatic solid tumors.

Pharmaceuticals – radio-isotopes

In addition to these osteoclast targeting agents, radio-isotopes may be used in the prevention of skeletal-related events. While beta-emitting particles (strontium-89 and samarium-153) may be used in the palliation of disease-related bony pain, they don’t have proven benefit in the prevention of SREs. In contrast, the alpha-emitting particle radium-223 has proven both palliative benefit as well as improvements in time to first SRE and overall survival. In the pivotal ALSYMPCA trial, 922 men with castrate-resistant prostate cancer and at least two symptomatic bony metastases who had either previously received or were unfit to receive docetaxel were randomized to radium-223 or placebo. Patients receiving radium-223 had significant improvements in time to first SRE (incremental benefit 5.2 months) as well as overall survival (incremental benefit in median survival of 2.8 months).2

Approach to preventing skeletal-related events

Philosophically, there are a number of times in the natural history of prostate cancer where a clinician may intervene to reduce the risk of prostate cancer related skeletal-related events. These relate to the complex interplay between prostate cancer and bone disease.

First, treatment may be directed at reducing or preventing fragility fractures due to prostate cancer-related therapy. In this disease space, lifestyle and nutrition-based interventions are paramount and are recommended by AUA guidelines as well as the National Comprehensive Cancer Network (NCCN). Pharmacologic interventions, including the use of osteoclast targeting therapies (bisphosphonates and RANK-ligand inhibitors), should be considered in this setting for patients at increased risk of fragility related fracture. Denosumab is FDA approved for this indication on the basis of the HALT 138 trial which found an increase in bone mineral density and a decrease in vertebral fractures among men receiving ADT for non-metastatic hormone-sensitive prostate cancer who receiving denosumab versus placebo.

Second, treatment may be directed at preventing bone metastases. In this disease space, bone-targeting agents including bisphosphonates and denosumab have been examined. The MRC PR04 trial demonstrated no benefit to clodronate in metastasis-free survival. Unfortunately, the Zometa 704 trial assessing the role of zoledronic acid in men with non-metastatic castration-resistant prostate cancer failed to accrue. However, the ZEUS trial accrued 1393 men with high-risk localized prostate cancer. Treatment with zoledronic acid failed to demonstrate an improvement in bone metastasis compared to placebo.7 The Denosumab Protocol 20050147 randomized men with high-risk nonmetastatic castrate-resistant prostate cancer to denosumab or placebo. Men receiving denosumab had prolonged metastasis-free survival (incremental benefit of 4.2 months).8

However, other approaches utilizing the suppression of the androgen axis in patients with non-metastatic castration-resistant prostate cancer (enzalutamide, apalutamide, and darolutamide) have demonstrated improvements in metastasis-free survival. While not a primary endpoint of these studies, it would be anticipated that delaying metastasis may improve skeletal-related events in these patients.

Third, we may seek to prevent skeletal-related events in men with known metastatic prostate cancer. This applies whether these men are in the castrate-sensitive or castrate-resistant disease space and it is here that the bulk of the evidence for prevention of SREs lies. In men with castrate-sensitive disease, CALGB 90202 demonstrated no improvement in skeletal-related events with the administration of zoledronic acid while castrate-sensitive rather than delayed initiation at the time of castration resistance.9 In contrast, there is significant evidence for the role of bone targeting agents in men with castrate-resistant disease. As previously mentioned, Zometa 039 demonstrated improvements in SREs for men receiving zoledronic acid compared with a placebo. Interestingly, CGP 032 and INT 05 failed to demonstrate a benefit to pamidronate. Thus, zoledronic acid is the only bisphosphonate approved in this space. The Denosumab Protocol 20050103 demonstrated an improvement in time to first SRE, as previously mentioned. Further, radio-isotope therapy using radium-223 was demonstrated in ALSYMPCA to improve time to first SRE in men with castrate-resistant prostate cancer and at least two symptomatic bony metastases.

Written by:  Zachary Klaassen, MD, MSc, Assistant Professor of Urology, Georgia Cancer Center, Augusta University/Medical College of Georgia, Twitter: @zklaassen_md

Published Date: February 2020

Written by: Zachary Klaassen, MD, MSc
References: 1. Morgans, Alicia K., and Matthew R. Smith. "Bone-targeted agents: preventing skeletal complications in prostate cancer." Urologic Clinics 39, no. 4 (2012): 533-546.
2. Parker, Christopher, S. Nilsson, Daniel Heinrich, Svein I. Helle, J. M. O'sullivan, Sophie D. Fosså, Aleš Chodacki et al. "Alpha emitter radium-223 and survival in metastatic prostate cancer." New England Journal of Medicine 369, no. 3 (2013): 213-223.
3. Watts, Nelson B., Robert A. Adler, John P. Bilezikian, Matthew T. Drake, Richard Eastell, Eric S. Orwoll, and Joel S. Finkelstein. "Osteoporosis in men: an Endocrine Society clinical practice guideline." The Journal of Clinical Endocrinology & Metabolism 97, no. 6 (2012): 1802-1822.
4. Lowrance, William T., Mohammad Hassan Murad, William K. Oh, David F. Jarrard, Matthew J. Resnick, and Michael S. Cookson. "Castration-resistant prostate cancer: AUA Guideline Amendment 2018." The Journal of urology 200, no. 6 (2018): 1264-1272.
5. Saad, Fred, Donald M. Gleason, Robin Murray, Simon Tchekmedyian, Peter Venner, Louis Lacombe, Joseph L. Chin, Jeferson J. Vinholes, J. Allen Goas, and Bee Chen. "A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma." Journal of the National Cancer Institute 94, no. 19 (2002): 1458-1468.
6. Fizazi, Karim, Michael Carducci, Matthew Smith, Ronaldo Damião, Janet Brown, Lawrence Karsh, Piotr Milecki et al. "Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study." The Lancet 377, no. 9768 (2011): 813-822.
7. Wirth, Manfred, Teuvo Tammela, Virgilio Cicalese, Francisco Gomez Veiga, Karl Delaere, Kurt Miller, Andrea Tubaro et al. "Prevention of bone metastases in patients with high-risk nonmetastatic prostate cancer treated with zoledronic acid: efficacy and safety results of the Zometa European Study (ZEUS)." European urology 67, no. 3 (2015): 482-491
8. Smith, Matthew R., Fred Saad, Robert Coleman, Neal Shore, Karim Fizazi, Bertrand Tombal, Kurt Miller et al. "Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial." The Lancet 379, no. 9810 (2012): 39-46.
9. Smith, Matthew R., Susan Halabi, Charles J. Ryan, Arif Hussain, Nicholas Vogelzang, Walter Stadler, Ralph J. Hauke et al. "Randomized controlled trial of early zoledronic acid in men with castration-sensitive prostate cancer and bone metastases: results of CALGB 90202 (alliance)." Journal of Clinical Oncology 32, no. 11 (2014): 1143.

Introduction: External Urinary Catheters

External urinary catheters (EUC) are used as collection devices or systems (referred in the UroToday reference center as external urine collection devices [EUCD]) for collecting and containing urine via tubing that relies on gravity to drain urine away from the penis or perineum into a drainage bag or suction that pulls urine into a container.
Written by: Diane K. Newman, DNP, ANP-BC, FAAN
References: 1. Beeson, Terrie, and Carmen Davis. "Urinary management with an external female collection device." Journal of Wound, Ostomy, and Continence Nursing 45, no. 2 (2018): 187.
2. Cottenden A, Fader M, Beeckma D, Buckley B, Kitson-Reynolds E, Moore K, Nishimura K, Ostaszkiewicz J, Watson J, Wilde M. (2017) "Management using continence products." In P. Abrams, L. Cardozo, S. Wagg, A. Wein. (Eds.). Incontinence: Proceedings from the 6th International Consultation on Incontinence (pp.2342-2346). ICUD ICS Publications
3. Deng, Donna Y. "Urologic Devices." In Clinical Application of Urologic Catheters, Devices and Products, pp. 173-220. Springer, Cham, 2018.
4. Fader, Mandy, Donna Bliss, Alan Cottenden, Katherine Moore, and Christine Norton. "Continence products: research priorities to improve the lives of people with urinary and/or fecal leakage." Neurourology and Urodynamics: Official Journal of the International Continence Society 29, no. 4 (2010): 640-644.
5. Geng, V., H. Cobussen-Boekhorst, H. Lurvink, I. Pearce, and S. Vahr. "Evidence-based guidelines for best practice in urological health care: male external catheters in adults urinary catheter management." Arnhem: European Association of Urology Nurses (2016).
6. Gould, C. V., C. A. Umscheid, R. K. Agarwal, G. Kuntz, and D. A. Pegues. "HICPAC." Guideline for prevention of catheter–associated urinary tract infections. CDC (2009). Infect Control Hosp Epidemiol. 2010 Apr;31(4):319-26. DOI: 10.1086/651091. PubMed PMID: 20156062.
7. Gray, Mikel, Claudia Skinner, and Wendy Kaler. "External collection devices as an alternative to the indwelling urinary catheter: evidence-based review and expert clinical panel deliberations." Journal of Wound, Ostomy, and Continence Nursing 43, no. 3 (2016): 301.
8. Lachance, Chantelle C., and Aleksandra Grobelna. "Management of Patients with Long-Term Indwelling Urinary Catheters: A Review of Guidelines." (2019).
9. Newman DK. (2017). "Devices, products, catheters, and catheter-associated urinary tract infections." In: Newman DK, Wyman JF, Welch VW, editors. Core Curriculum for Urologic Nursing. 1st ed. Pitman (NJ): Society of Urologic Nurses and Associates, Inc; 439-66.
10. Newman, Diane K., and Alan J. Wein. "External Catheter Collection Systems." In Clinical Application of Urologic Catheters, Devices and Products, pp. 79-103. Springer, Cham, 2018.

Expanding Treatment Options in Non-metastatic Castrate-resistant Prostate Cancer

Prostate cancer (PCa) is the second most common form of cancer diagnosed in US men. It represents 19% of newly diagnosed cancers, and the third leading cause of cancer death, accounting for an estimated 39,430 deaths in 2018.1 
Written by: Hanan Goldberg MD, Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA
References:
  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. Jan 2018;68(1):7-30.
  2. Trapasso JG, deKernion JB, Smith RB, Dorey F. The incidence and significance of detectable levels of serum prostate specific antigen after radical prostatectomy. J Urol. Nov 1994;152(5 Pt 2):1821-1825.
  3. Tombal B, Miller K, Boccon-Gibod L, et al. Additional analysis of the secondary end point of biochemical recurrence rate in a phase 3 trial (CS21) comparing degarelix 80 mg versus leuprolide in prostate cancer patients segmented by baseline characteristics. Eur Urol. May 2010;57(5):836-842.
  4. Karantanos T, Evans CP, Tombal B, Thompson TC, Montironi R, Isaacs WB. Understanding the mechanisms of androgen deprivation resistance in prostate cancer at the molecular level. Eur Urol. Mar 2015;67(3):470-479.
  5. Saad F, Hotte SJ. Guidelines for the management of castrate-resistant prostate cancer. Canadian Urological Association journal = Journal de l'Association des urologues du Canada. 2010;4(6):380-384.
  6. Alpajaro SIR, Harris JAK, Evans CP. Non-metastatic castration resistant prostate cancer: a review of current and emerging medical therapies. Prostate Cancer Prostatic Dis. Mar 2019;22(1):16-23.
  7. Chandrasekar T, Yang JC, Gao AC, Evans CP. Mechanisms of resistance in castration-resistant prostate cancer (CRPC). Transl Androl Urol. Jun 2015;4(3):365-380.
  8. Sharifi N, Dahut WL, Steinberg SM, et al. A retrospective study of the time to clinical endpoints for advanced prostate cancer. BJU Int. Nov 2005;96(7):985-989.
  9. Macomson B, Lin JH, Tunceli O, et al. Time to metastasis or death in non-metastatic castrate resistant prostate cancer (nmCRPC) patients by National Comprehensive Cancer Network (NCCN) risk groups. Journal of Clinical Oncology. 2017;35(15_suppl):5027-5027.
  10. Scher HI, Solo K, Valant J, Todd MB, Mehra M. Prevalence of Prostate Cancer Clinical States and Mortality in the United States: Estimates Using a Dynamic Progression Model. PLoS One. 2015;10(10):e0139440.
  11. Lowrance WT, Murad MH, Oh WK, Jarrard DF, Resnick MJ, Cookson MS. Castration-Resistant Prostate Cancer: AUA Guideline Amendment 2018. J Urol. Dec 2018;200(6):1264-1272.
  12. Scher HI, Morris MJ, Stadler WM, et al. Trial Design and Objectives for Castration-Resistant Prostate Cancer: Updated Recommendations From the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol. Apr 20 2016;34(12):1402-1418.
  13. Liede A, Arellano J, Hechmati G, Bennett B, Wong S. International prevalence of nonmetastatic (M0) castration-resistant prostate cancer (CRPC). Journal of Clinical Oncology. 2013;31(15_suppl):e16052-e16052.
  14. Smith MR, Saad F, Chowdhury S, et al. Apalutamide Treatment and Metastasis-free Survival in Prostate Cancer. New England Journal of Medicine. 2018;378(15):1408-1418.
  15. Virgo KS, Rumble RB, Singer EA. Second-Line Hormonal Therapy for Men With Chemotherapy-Naive Castration-Resistant Prostate Cancer: American Society of Clinical Oncology Provisional Clinical Opinion Summary. J Oncol Pract. Jul 2017;13(7):459-461.
  16. Crawford ED, Stone NN, Yu EY, et al. Challenges and recommendations for early identification of metastatic disease in prostate cancer. Urology. Mar 2014;83(3):664-669.
  17. Sartor AO, Tangen CM, Hussain MH, et al. Antiandrogen withdrawal in castrate-refractory prostate cancer: a Southwest Oncology Group trial (SWOG 9426). Cancer. Jun 2008;112(11):2393-2400.
  18. Murray NP, Reyes E, Tapia P, Badínez L, Orellana N. Differential expression of matrix metalloproteinase-2 expression in disseminated tumor cells and micrometastasis in bone marrow of patients with nonmetastatic and metastatic prostate cancer: theoretical considerations and clinical implications-an immunocytochemical study. Bone marrow research. 2012;2012:259351-259351.
  19. Eiber M, Maurer T, Souvatzoglou M, et al. Evaluation of Hybrid (6)(8)Ga-PSMA Ligand PET/CT in 248 Patients with Biochemical Recurrence After Radical Prostatectomy. J Nucl Med. May 2015;56(5):668-674.
  20. Morigi JJ, Stricker PD, van Leeuwen PJ, et al. Prospective Comparison of 18F-Fluoromethylcholine Versus 68Ga-PSMA PET/CT in Prostate Cancer Patients Who Have Rising PSA After Curative Treatment and Are Being Considered for Targeted Therapy. J Nucl Med. Aug 2015;56(8):1185-1190.
  21. Umbehr MH, Muntener M, Hany T, Sulser T, Bachmann LM. The role of 11C-choline and 18F-fluorocholine positron emission tomography (PET) and PET/CT in prostate cancer: a systematic review and meta-analysis. Eur Urol. Jul 2013;64(1):106-117.
  22. Geynisman DM, Plimack ER, Zibelman M. Second-generation Androgen Receptor-targeted Therapies in Nonmetastatic Castration-resistant Prostate Cancer: Effective Early Intervention or Intervening Too Early? Eur Urol. Dec 2016;70(6):971-973.
  23. Taylor CD, Elson P, Trump DL. Importance of continued testicular suppression in hormone-refractory prostate cancer. J Clin Oncol. Nov 1993;11(11):2167-2172.
  24. Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol. Nov 10 2005;23(32):8253-8261.
  25. Shah RB, Mehra R, Chinnaiyan AM, et al. Androgen-independent prostate cancer is a heterogeneous group of diseases: lessons from a rapid autopsy program. Cancer Res. Dec 15 2004;64(24):9209-9216.
  26. Maximum androgen blockade in advanced prostate cancer: an overview of the randomised trials. Prostate Cancer Trialists' Collaborative Group. Lancet. Apr 29 2000;355(9214):1491-1498.
  27. Schweizer MT, Zhou XC, Wang H, et al. Metastasis-free survival is associated with overall survival in men with PSA-recurrent prostate cancer treated with deferred androgen deprivation therapy. Ann Oncol. Nov 2013;24(11):2881-2886.
  28. Schroder FH, Tombal B, Miller K, et al. Changes in alkaline phosphatase levels in patients with prostate cancer receiving degarelix or leuprolide: results from a 12-month, comparative, phase III study. BJU Int. Jul 2010;106(2):182-187.
  29. Tran C, Ouk S, Clegg NJ, et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science. May 8 2009;324(5928):787-790.
  30. Hussain M, Fizazi K, Saad F, et al. Enzalutamide in Men with Nonmetastatic, Castration-Resistant Prostate Cancer. New England Journal of Medicine. 2018;378(26):2465-2474.
  31. Fizazi K, Shore N, Tammela TL, et al. Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer. New England Journal of Medicine. 2019;380(13):1235-1246.
  32. El-Amm J, Aragon-Ching JB. The Current Landscape of Treatment in Non-Metastatic Castration-Resistant Prostate Cancer. 2019;13:1179554919833927.
  33. Clegg NJ, Wongvipat J, Joseph JD, et al. ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Res. Mar 15 2012;72(6):1494-1503.
  34. Scher HI, Fizazi K, Saad F, et al. Increased Survival with Enzalutamide in Prostate Cancer after Chemotherapy. New England Journal of Medicine. 2012;367(13):1187-1197.
  35. Beer TM, Armstrong AJ, Rathkopf DE, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. Jul 31 2014;371(5):424-433.
  36. Xie W, Regan MM, Buyse M, et al. Metastasis-Free Survival Is a Strong Surrogate of Overall Survival in Localized Prostate Cancer. J Clin Oncol. Sep 20 2017;35(27):3097-3104.
  37. Shore ND. Darolutamide (ODM-201) for the treatment of prostate cancer. Expert Opin Pharmacother. Jun 2017;18(9):945-952.
  38. Smith MR, Saad F, Oudard S, et al. Denosumab and bone metastasis-free survival in men with nonmetastatic castration-resistant prostate cancer: exploratory analyses by baseline prostate-specific antigen doubling time. J Clin Oncol. Oct 20 2013;31(30):3800-3806.
  39. Nelson JB, Love W, Chin JL, et al. Phase 3, randomized, controlled trial of atrasentan in patients with nonmetastatic, hormone-refractory prostate cancer. Cancer. Nov 1 2008;113(9):2478-2487.
  40. Miller K, Moul JW, Gleave M, et al. Phase III, randomized, placebo-controlled study of once-daily oral zibotentan (ZD4054) in patients with non-metastatic castration-resistant prostate cancer. Prostate Cancer Prostatic Dis. Jun 2013;16(2):187-192.
  41. Beinart G, Rini BI, Weinberg V, Small EJ. Antigen-presenting cells 8015 (Provenge) in patients with androgen-dependent, biochemically relapsed prostate cancer. Clin Prostate Cancer. Jun 2005;4(1):55-60.
  42. Madan RA, Gulley JL, Schlom J, et al. Analysis of overall survival in patients with nonmetastatic castration-resistant prostate cancer treated with vaccine, nilutamide, and combination therapy. Clin Cancer Res. Jul 15 2008;14(14):4526-4531.
  43. Ogita S, Tejwani S, Heilbrun L, et al. Pilot Phase II Trial of Bevacizumab Monotherapy in Nonmetastatic Castrate-Resistant Prostate Cancer. ISRN oncology. 2012;2012:242850-242850.
  44. Ryan CJ, Crawford ED, Shore ND, et al. The IMAAGEN Study: Effect of Abiraterone Acetate and Prednisone on Prostate Specific Antigen and Radiographic Disease Progression in Patients with Nonmetastatic Castration Resistant Prostate Cancer. J Urol. Aug 2018;200(2):344-352.
  45. Hussain M, Corn PG, Michaelson MD, et al. Phase II study of single-agent orteronel (TAK-700) in patients with nonmetastatic castration-resistant prostate cancer and rising prostate-specific antigen. Clin Cancer Res. Aug 15 2014;20(16):4218-4227.
  46. Fizazi K, Jones R, Oudard S, et al. Phase III, randomized, double-blind, multicenter trial comparing orteronel (TAK-700) plus prednisone with placebo plus prednisone in patients with metastatic castration-resistant prostate cancer that has progressed during or after docetaxel-based therapy: ELM-PC 5. J Clin Oncol. Mar 1 2015;33(7):723-731.