Sunday, October 14, 2018

#ProstateCancer News 2018-10


Prostate Cancer News of interest for October, 2018
Contents


The Next IPCSG Meeting


Saturday October 15, 2018 10:00 AM
Member Panel      Personal PCa Stories

--------------------------------------------------------------------------------------------------------------------------------------

Nutrition, ADT, and metastatic prostate cancer

Posted on October 8, 2018

Most men who are taking androgen deprivation therapy (ADT) for metastatic prostate cancer feel the need to “do something” about their diet in order to (a) further help to control the risk for progression of their cancer and (b) help to cope with the side effects of ADT.
Another paper just published in Prostate Cancer and Prostatic Diseases this month is a review by Barnes et al. on whether current nutrition care guidelines for men with prostate cancer who are being treated with ADT are really based on good enough evidence.
The authors claim to have been able to find just 16 articles on this topic that met the inclusion criteria for their review, and they summarize their findings as follows:
·         Each of the 16 articles offered distinct sets of dietary interventions designed to manage the side effects of ADT.
·         12/16 articles combined nutritional guidelines with physical activity and/or medications and/or counseling.
·         4/16 articles dealt exclusively with the impacts of diet alone, and among these four articles
o   Three articles measured changes to participants’ dietary intake and influence on ADT side effects.
o   One article showed daily caffeinated beverages improved cancer-related fatigue.
o   Two articles showed no impact of isoflavone supplementation on hot flashes, quality of life, body mass index, or blood lipids.
·         Among the 16 articles altogether
o   Dietary intake and compliance was poorly reported (and thus provided limited knowledge of acceptability and feasibility for dietary interventions).
o   Information on the nutrition care practices and views of clinicians treating men for prostate cancer is limited.
o   No articles measured the impact of diet on long-term side effects of ADT.
o   The methodological quality of the papers ranged from weak to strong.
Barnes et al. conclude that:
·         Current evidence for dietary interventions to mitigate ADT side effects is limited. Further investigations are warranted to explore the impact of changes in dietary intake on ADT side effects before practice guidelines can be considered.
We have considerable sympathy with the findings of Barnes et al. In general, our knowledge of what really “works” from a dietary and a nutritional perspective to optimize the quality of life of men with prostate cancer and of men with prostate cancer who are on ADT is poor.
While we know what is a really bad idea (e.g., diets high in red meat and carbohydrates and animal fats) and we know that some men believe they do really well on diets that are almost entirely vegetarian or vegan, the question of how well any of these diets actually work for large numbers of men has yet to be answered in any really well conducted studies.
Thus our advice to most men at the present time generally takes three forms:
·         Eat a well balanced diet that is significantly lower in red meat, carbohydrates, animal fats, and sugars than the average US diet today.
·         Eat more cruciferous vegetables (things like broccoli, cauliflower, cabbage, and brussels sprouts), nuts, and roughage.
·         Exercise regularly and work up a good sweat if you can when you do.
We know these things are good for the heart and for other bodily functions too, and there is plenty of evidence to suggest they are also good for people with cancers of many types.
However, it would be really nice to know if specific diets and supplements had real, definable benefits for men on ADT. Many men swear by the value of soybean-based products as a way to down-grade the impact of ADT on hot flashes, but whether this is really true for most men — based on a well-controlled and randomized trial — remains utterly unknown.

--------------------------------------------------------------------------------------------------------------------------------------

Personalized Medicine in Prostate Cancer: Now More Precise

Raoul S. Concepcion, MD, FACS
Over the past decade, we have witnessed an explosion of molecular testing in the urology world that will help us better diagnose and manage prostate cancer across the spectrum of the disease. All of us recognize that we have fallen behind our colleagues who treat other tumor types and for whom routine lab and tissue testing is the requirement or norm for optimal therapeutic choices. However, we are making progress.
We know that prostate cancer is an endocrine disease and the androgen receptor (AR) plays a major role in disease progression, even when serum testosterone is at castration levels of the hormone. Androgen deprivation therapy (ADT) continues to be the foundation of treatment on which all other oncolytics are layered as the patient begins to progress. However, treatment pressure selection and tumor heterogeneity will result in cellular mutagenesis that makes ultimately eradicating the disease a daunting task. In patients with metastatic castration resistant prostate cancer, a number of these mutations, which will inevitably confer resistance to therapy, have been discovered. For example, prostate cancer cells that have been long exposed to ADT and are now starved of their fuel for survival can produce their own ligands to survive. This is comparable to a car that could produce its own gasoline (or electricity)—very nice if you are the automobile owner—but the reality is not so desirable for the patient with cancer. AR, the very target of ADT, can undergo genetic alterations that will result in downstream changes leading to resistance and cell survival.
Fortunately, we are in the early stages of developing biomolecular markers that will help the clinician to choose better therapies to try to stay ahead of these mutations. The ability to measure circulating tumor cells has led to the development and marketing of commercial assays to detect these aberrations—AR splice variants—that may be predictive of what agent should be used next in the treatment paradigm. In addition, patients who harbor homozygous mutations of inherited DNA repair genes, most notably BRCA2, may in fact respond to PARP inhibition therapy. Also, DNA sequencing to test for these gene mutations can now be clinically implemented in the appropriate patient.
Thus, just as it is difficult to keep track of all the FDA-approved therapies, their mechanisms of action, adverse event profiles, and current approval status, we are additionally challenged to stay up-to-date with the molecular tests that may ultimately be required to determine when and where to use these drugs in the appropriate sequence for each patient. The vernacular commonly used in the press is the “era of personalized medicine.” I will argue that the art of medicine has always been personalized. We as providers do not adhere to a single agent, from an antibiotic to an antihypertensive to an oncolytic, for all patients. All of us consider the patient’s medical history and comorbidities and a host of other factors to best determine which agent to prescribe in hopes of achieving the best outcome. Now, given the complexity in understanding these malignancies and their ability to undergo evolutionary mutations to survive, we must clearly understand how to accurately test and target these dynamics to stay ahead of the changing tumor environment. Personalized yet ever more precise at the tumor level—precision medicine. This is how all of us will need to practice moving forward to continue making strides in advancing cancer care.

--------------------------------------------------------------------------------------------------------------------------------------

What to Expect When Active Surveillance Leads to Prostatectomy | Medpage Today: High-risk features in 39% of prostate specimens at surgery

Oncology/Hematology > Prostate Cancer
    by Charles Bankhead, Senior Associate Editor, MedPage Today
    September 21, 2018
A high proportion of men who entered active surveillance for early prostate cancer had one or more high-risk disease characteristics when they subsequently had radical prostatectomy, a Swedish study showed.
Medical records showed that 52 of 132 men had at least one adverse pathology feature at radical prostatectomy. All the men initially opted for active surveillance, and the median time from enrollment to surgery was 1.9 years.
Adverse pathology findings included Gleason score >3 + 4, extraprostatic extension, positive surgical margins, seminal vesicle invasion, and lymph node involvement, as reported in the Journal of Urology.
"Our findings can be used to counsel patients on AS (active surveillance) regarding what to expect in those who progression during surveillance and undergo deferred radical prostatectomy, as 85% of the cohort had objective signs of progression triggering treatment, including upgrading or increased cancer volume or PSA (prostate-specific antigen)," Rebecka Arnsrud Godtman, MD, of the University of Goteborg and Sahlgrenska University Hospital, and co-authors wrote.
"The results underscore the need to determine better methods of risk classification and identify progression during AS to select the best treatment strategy for each individual patient at each phase of the disease."
Another study in the same issue of the journal showed positive results with a minimally invasive technique that dramatically reduced the frequency of progression to radical prostatectomy among men on active surveillance. The 2-year rate of conversion was 7% among men who had tissue ablation with vascular targeted phototherapy (VTP) prior to beginning active surveillance versus 32% for those who had no treatment prior to starting surveillance. At 4 years, the prostatectomy rates were 24% in the VTP group and 53% in the surveillance group, reported Inderbir Gill, MD, of the University of Southern California in Los Angeles, and colleagues.
"Absolutely," Gill told MedPage Today when asked whether VTP might address the progression issue observed in the Swedish study. "What we're showing is that progression to radical prostatectomy occurs less often for [VTP] than with active surveillance -- about 30% less often at 4 years.
"Conversion to radical prostatectomy is primarily due to progression of the cancer," he added. "[VTP] arrests progression and decreases the need for radical therapy up to 4 years now, and we already have 5-year data in the works."
The findings added to those that Gill reported earlier this year at the American Urological Association annual meeting. The findings came from a multicenter randomized trial of the TOOKAD VTP system (Steba Biotech), which employs low-level laser energy and light-activated padeliporfin di-potassium to induce vascular necrosis and tissue ablation. Not yet available in the U.S., the TOOKAD device has regulatory approval in Mexico, Israel, and most of the European Union.
Although a growing number of men with early prostate cancer opt for active surveillance, half the patients eventually have radical therapy (surgery or definitive radiation therapy). Combined with the proportion of men who have upfront surgery or radiotherapy, about 70% of men still undergo definitive treatment for early prostate cancer, despite the emphasis on active surveillance to reduce overtreatment of low-risk disease, Gill noted.
Inherent to the underlying rationale for active surveillance is the premise that the time spent in surveillance will not jeopardize the chance of curing the disease, Godtman and coauthors noted in their introduction. As a result, meticulous selection and follow-up criteria are essential components of any active surveillance program.
The primary objectives of the Swedish study were to determine long-term outcomes of men who deferred surgery in favor of active surveillance and to determine how often tumors removed during radical prostatectomy are missed during interval biopsies. Such information could inform development of active surveillance programs and counseling patients about active surveillance for early prostate cancer, the authors added.
Study Details
The analysis involved 132 men with screen-detected prostate cancer and who entered active surveillance and subsequently underwent radical prostatectomy during 1995 through 2014. Follow-up in the cohort continued to May 2017. During active surveillance, patients had PSA tests every 3 to 6 months and follow-up biopsies every 2 to 4 years. Triggers for radical prostatectomy included rising PSA, increased cancer grade or stage, and patient preference.
The study population had a median age of 64 and a median PSA level of 4.1 ng/mL. At diagnosis, 73 of the 132 men (55%) had very low-risk prostate cancer, 30% had low-risk disease, and 14% had intermediate-risk disease characteristics.
The cohort had a median interval from prostate cancer diagnosis to radical prostatectomy of 1.9 years and median postoperative follow-up of 10.9 years. Pathology reports for prostatectomy specimens showed that 39% of the patients had at at least one adverse disease characteristic at surgery.
On the basis of the pathologic features observed at surgery, the tumor was not identified by the diagnostic biopsy in 29% of all 132 cases or by the repeat biopsy that preceded radical prostatectomy in 21% of 105 cases, the authors reported. The study population had a 10-year PSA relapse-free survival rate of 79.5%.
The study by Godtman et al was supported by the Swedish Cancer Society. The authors reported having no relevant relationships with industry.
The study by Gill et al was supported by the National Cancer Institute, the National Institute of Health Research (England), several private foundations, and Steba Biotech. The authors reported having no relevant relationships with industry.

--------------------------------------------------------------------------------------------------------------------------------------

Prostate Cancer Paradigm Progresses, But Unanswered Questions Remain

Brandon Scalea
Randy F. Sweis, MD
Large phase III studies, such as the CHAARTED and STAMPEDE trials, have helped to significantly improved outcomes for men with prostate cancer, but unanswered questions remain, including what role immunotherapy could play in the treatment paradigm, said Randy F. Sweis, MD.
Long-term survival data from CHAARTED demonstrated prolonged overall survival (OS) benefit with docetaxel when added to androgen deprivation therapy (ADT) for patients with high-volume metastatic hormone-sensitive prostate cancer. At a median follow-up of 53.7 months, median OS was 57.6 months for the docetaxel/ADT arm versus 47.2 months for ADT alone.1
The STAMPEDE trial showed that in patients with locally advanced disease, ADT plus abiraterone acetate (Zytiga) and prednisone was associated with higher rates of OS and failure-free survival than ADT alone.2
Sweis, an instructor of medicine at the University of Chicago, said ongoing trials are testing immunotherapy in patients with prostate cancer. For example, the CheckMate-9KD study will look at nivolumab (Opdivo) combined with either a PARP inhibitor, androgen receptor therapy, or docetaxel in patients with metastatic castration-resistant disease.
In an interview during the 2018 OncLive® State of the Science Summit™, Sweis discussed recent progress and next steps in the prostate cancer paradigm.
OncLive: How has the treatment landscape of prostate cancer evolved in recent years?
Sweis: Prostate cancer is an interesting disease because numerous therapies have been FDA approved over the last few years. One of the areas in which it is most quickly evolving is the sequencing and timing of therapies. This has been an area of somewhat confusion, but we have also been able to make a significant impact on outcomes. Patients can live longer and have more time being in control of their cancer.
We are using these therapies that were initially approved in the metastatic castration-resistant setting early on. A lot of these are secondary androgen-targeting agents, such as enzalutamide (Xtandi) and apalutamide (Erleada). Abiraterone acetate and chemotherapy regimens, such as docetaxel—and these drugs have been around for quite a while—are being utilized in this space effectively. We're using them earlier. We're treating patients a lot more aggressively than we were 5 years ago. The duration of control of the disease is being extended.
We are seeing this in large international studies such as CHAARTED or the STAMPEDE trial. That is where the most change is occurring. There are some efforts investigating things like immunotherapy in prostate cancer. That's been a challenging area, but there are a lot of people working on it and it has some potential in this field. We've seen a couple responders with immunotherapy; we just need a better understanding of the biology. This is an area where we can see some breakthrough in the next 5 years.
Are there any indications as to a patient population who might best respond to immunotherapy?
We don't know quite yet, especially in prostate cancer. How to select patients for this type of treatment is tricky, and it's something we definitely need more data for. We will hopefully see that in the next few years. It's an area of ongoing investigation.
We have some trials at our institution looking at combining a PARP inhibitor with an anti–PD-1 agent. This is a novel approach. There are others looking at the combination of ipilimumab (Yervoy) and nivolumab, which we've seen have promise in other cancer types. We have some hope here, and we'll see how it plays out. We've seen some responses, as I said before. It's just a matter of figuring out the right combination.
What are the remaining questions with regard to sequencing?
We have a lot of drugs now, especially those targeting the testosterone and androgen pathway. Several of them have shown success, and now there is a question of which one is optimal. Could these be combined with another therapy? Also, there is the chemotherapy question. Docetaxel given in the metastatic castrate-sensitive setting has been shown to improve survival. However, so has abiraterone. Some patients are receiving either one. This is going to be a big question. Which therapies should we be using upfront? We have to test them head to head.
The other question is of radium-223 dichloride (Xofigo). It's an infusion that targets bone metastases, and it's shown efficacy in castration-resistant patients. We have to see where this really fits in.
References
1.    Kyriakopoulos C, Chen Y, Carducci M, et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED Trial. J Clin Oncol. 2018;36(11):1080-1087. doi: 10.1200/JCO.2017.75.3657.
2.    James N, de Bono J, Spears M, et al. Abiraterone for prostate cancer not previously treated with hormone therapy. N Engl J Med. 2017;377:338-351. doi: 10.1056/NEJMoa1702900.

--------------------------------------------------------------------------------------------------------------------------------------

ANO7 Gene Linked to Aggressive Prostate Cancer | Cancer Network

John Schieszer Sep 20, 2018
The prostate-specific (anoctamin 7) ANO7 gene may be a key player in the development of aggressive prostate cancer, found a new European study published in the International Journal of Cancer. By elevating the expression of ANO7, a potential prostate cancer susceptibility gene, it may be possible to predict disease severity and outcome, according to the researchers.
“We have hypothesized for a long time that, at the molecular level, there's probably a continuum of diseases that we now call prostate cancer. Prostate cancer has a wide spectrum of clinical behavior that ranges from decades of indolence to rapid metastatic progression and lethality. The biggest treatment challenge is determination of aggressive cancers already in early phases of the disease,” said study investigator Johanna Schleutker, PhD, professor of medical genetics, Institute of Biomedicine, University of Turku/Division of Laboratory, Turku University Hospital, Turku, Finland.
Schleutker and her colleagues sequenced ANO7 in castration-resistant tumors together with samples from unselected prostate cancer patients and unaffected men. The team uncovered two pathogenic variants which were then genotyped in 1,769 men with prostate cancer and 1,711 men without prostate cancer. Different databases along with Swedish and Norwegian cohors were used to help validate the findings.
Variant rs77559646 was associated with increased risk (OR 1.40; 95% CI, 1.09-1.78; P = .009) and with aggressive prostate cancer (Genotype test, P = .04). In addition, rs148609049 was not associated with prostate cancer risk, but was related to shorter survival (HR, 1.56; 95% CI, 1.03-2.36). Overall, high ANO7 expression was independently linked to poor survival (HR, 18.4; 95% CI, 1.43-237).
Currently, there is no clear way to diagnose aggressive prostate cancer at an early stage. However, genetic mutations such as those revealed in this study could lead to the development of accurate diagnostic tests. “Obviously, more research is needed to understand the mechanisms behind our findings. However, it is very likely that genomics will become an important tool for decision-making in prostate cancer treatment,” Schleutker told Cancer Network. Genetic testing for ANO7 could provide new opportunities for precision oncology in prostate cancer, she added.
The researchers noted that although this study involved a large population, it is limited by the fact that it primarily consists of Caucasians from Northern Europe. Further research involving other demographics is warranted to validate the findings, they noted.
William Catalona, MD, a professor of urology at Northwestern Medicine Feinberg School of Medicine, Chicago, said this study and others are providing an increased understanding of the genomics of prostate cancer. For instance, germline variants associated with deficiencies of DNA repair have been shown to increase the susceptibility to prostate cancer and its aggressiveness.
“This information has helped define distinct genomic pathways with different clinical outcomes [that] respond to different treatments,” Catalona told Cancer Network. “This excellent study performed by a highly-respected, experienced research team has identified a relatively rare genetic variant that could be inexpensively genotyped as part of a biomarker panel that, along with other parameters, could help distinguish men with aggressive prostate cancer. The authors’ caveat about validation in non-northern European white populations is important,” he confirmed.

--------------------------------------------------------------------------------------------------------------------------------------

Complications of Androgen Deprivation Therapy in Men With Prostate Cancer

    Tejas Patil, MDBrandon Bernard, MD, MPH
Sep 18, 2018 Volume: 32 Issue:  9 Prostate Cancer, Genitourinary Cancers, Oncology Journal Oncology (Williston Park). 32(9):470-4, CV3.
Table. Major Side Effects Associated With Androgen Deprivation Therapy (ADT)
Abstract / Synopsis:
The standard treatment for men with metastatic prostate cancer is androgen deprivation therapy (ADT). This therapy is associated with a multitude of side effects that can impact quality of life. These include vasomotor complications (in particular, hot flushes), sexual dysfunction and gynecomastia, osteoporosis, metabolic syndrome, and depression. Additionally, ADT has been associated with neurocognitive deficits, thromboembolic disease, and cardiovascular disease, although the data regarding the latter associations are mixed. This article summarizes the key side effects associated with ADT and discusses strategies to optimize management.
Introduction
Metastatic prostate cancer remains a global health challenge. Despite improvements in anticancer therapies, management of the disease is often a struggle to maintain a balance between preserving quality of life and extending survival. Charles Huggins and Andrew Schally laid the groundwork for establishing the critical role of antiandrogen therapies in the management of metastatic prostate cancer.[1] Initially, androgen deprivation therapy (ADT) was achieved via orchiectomy, but by the mid-1980s, the approval of gonadotropin-releasing hormone (GnRH) agonists allowed for medical ADT.[2] Intermittent ADT represents an option for patients with biochemically relapsed prostate cancer, but continuous ADT is the standard initial treatment for metastatic disease. While prolonged disease control can be achieved with this approach, use of ADT is associated with a wide range of side effects (Table) that may significantly reduce quality of life. In this article, we summarize the key side effects associated with ADT and highlight strategies to mitigate them.
Vasomotor Complications
Hot flushes are one of the most common and bothersome side effects associated with ADT. Up to 80% of patients undergoing treatment with GnRH agonists will experience hot flushes, with 27% of these patients reporting this symptom as the most significant adverse side effect.[3] Hot flushes are described as an intense feeling of warmth, occasionally associated with nausea and night sweats. Many patients will achieve some improvement in symptom severity as their bodies acclimate to the low testosterone state, although some patients will not acclimate. Therapeutic management is largely extrapolated from studies in breast cancer patients receiving tamoxifen.[4] Randomized clinical trials have demonstrated efficacy of gabapentin and venlafaxine; these agents may be used for this off-label indication in men receiving ADT.[5,6] Low-dose estrogen and progestins can also be considered,[5] but the benefits of this option need to be weighed against the thromboembolic risk associated with estrogen use.[7] Additionally, acupuncture has been shown to be of potential benefit.[8] Conservative strategies to abort hot flushes include sleeping with an open window or drinking cold beverages. Lastly, regular exercise can help decrease frequency and severity of hot flushes; given the benefit of exercise for other ADT side effects and overall health, this recommendation is strongly encouraged. Overall, a stepwise approach is suggested for hot flush management, including exercise, conservative measures, use of venlafaxine or gabapentin, and lastly use of estrogen or progestins as needed, while weighing the potential risks vs benefits of each treatment option.
Sexual Dysfunction and Gynecomastia
Several studies have shown that treatment for clinically localized prostate cancer involving either radical prostatectomy, brachytherapy, or external beam radiotherapy (EBRT) can result in long-term erectile dysfunction.[9,10] Patients receiving neoadjuvant ADT with EBRT were found to have decreased frequency of erection, decreased overall sexual function, and an increase in frequency of hot flushes.[11] In addition, loss of libido is a major consequence of ADT. The degree of erectile dysfunction for patients on ADT is impacted by pretreatment sexual function, as well as by changes in libido; in patients previously able to attain an erection, phosphodiesterase 5 inhibitors, intracavernosal injection therapy, vacuum-assisted devices, or other topical agents may be used. Given the complex interplay between physiology, psychology, stress, and emotion involved in sexual function, a referral to a psychology or counseling service with a focus on sexual health is recommended for interested patients and their partners.
Gynecomastia may arise in conditions of unopposed estrogen activity and can be distressing for cosmetic reasons or because of pain. It may occur with ADT; however, it is more problematic with antiandrogens (specifically at higher doses) due to the buildup of androgens and their consequent peripheral conversion to estrogen in breast tissue. Tamoxifen and breast irradiation are two treatment options; the former is more beneficial as primary prevention, whereas the latter may be of greater use following established gynecomastia.[12,13]
Bone Health
ADT is associated with significant changes in bone. Multiple studies have demonstrated that bone mineral density (BMD) is significantly reduced in men receiving ADT compared with controls.[14] The risk of fracture increases with use of ADT and appears to be dose dependent.[15] Prevention of skeletal-related events (SREs) has been evaluated in multiple randomized clinical trials for patients with nonmetastatic castration-sensitive prostate cancer (CSPC). One study demonstrated a significant decrease in BMD in men receiving ADT alone, while there was no significant change in those who received ADT plus pamidronate.[16] In castration-resistant prostate cancer (CRPC) metastatic to bone, a randomized placebo-controlled trial found that zoledronic acid reduced SREs.[17] Another phase III randomized clinical trial found denosumab to be more effective than zoledronic acid in preventing SREs.[18] Interestingly, in contrast to men with metastatic CRPC, no benefit was seen from osteoclast inhibition in men with CSPC metastatic to bone.[19]
Men who are about to start ADT should be evaluated for risk of osteoporosis, prior fractures, alcohol use, and prior (or current) glucocorticoid use. Serum vitamin D and baseline BMD may be checked to see if bisphosphonates are indicated, with periodic re-evaluation of BMD while on ADT. For all patients starting ADT, it is recommended that they supplement their diet with at least 1,000-mg oral calcium daily and 1,000 IU of vitamin D in an effort to preserve bone integrity and minimize losses; a referral to a registered dietician may further aid in refining both dietary and supplement recommendations. Lastly, an exercise program that includes resistance training and/or weight-bearing exercise is encouraged to further maintain bone strength.
Metabolic Syndrome
A variety of metabolic changes have been observed with ADT, including weight gain, dyslipidemia, hyperglycemia, and sarcopenia. Weight gain, specifically, is a well-recognized side effect of ADT.[20-22] Retrospective studies have found that men on ADT for nonmetastatic prostate cancer had a median weight gain of 1 to 2 kg after 1 year of treatment.[22] Similar changes in weight have been noted in the metastatic setting.[23-25] When more dramatic, weight gain can have major consequences in terms of both mental and cardiovascular health. A referral to a registered dietician may help with improving diet, while engagement in a structured and formal exercise program can help with reducing or maintaining weight. Many cancer centers offer such programs, and these should be suggested to interested or at-risk patients.
In addition, multiple studies have noted dyslipidemia and increased risk for metabolic syndrome with use of ADT.[26,27] Extrapolating that these changes are associated with increases in cardiovascular risk remains controversial. Loss of lean muscle mass and sarcopenic obesity have been described in several studies. A study of 39 men receiving ADT using CT measurements of the rectus, femoris, sartorius, and quadriceps muscles noted decreases in the cross-sectional area of these muscles.[28] Given the importance of these muscles in gait and balance, studies have explored the association between ADT use and fall risk. One study noted a higher rate of falls in persons treated with ADT, with apparent persistent risk even after ADT is discontinued.[29] Thus, judicious use of ADT and careful evaluation of frail patients and those at risk for complications are recommended when the need for treatment is questionable (eg, biochemical relapse with slowly rising prostate-specific antigen [PSA] levels).
Neurocognitive Changes
Emerging data show that men with prostate cancer receiving ADT are at risk for a spectrum of neurocognitive complications, such as depression, memory difficulties, and fatigue.[30] In a population-based cross-sectional study of elderly men, it was found that lower serum testosterone levels were associated with decreased performance on the Mini-Mental State Examination (MMSE), the Trail Making Test B, and the Digit Symbol Substitution cognitive test. A major confounding variable in this study was age, since more than half of all cases of dementia occur in men > 65 years of age. This was addressed in a large cohort study that found a statistically significant association between use of ADT and risk of dementia.[31] Sensitivity analyses found that use of ADT for longer than 12 months was associated with the greatest absolute risk of dementia.
Numerous prospective studies examining the impact of ADT on cognitive function have been performed, with mixed and conflicting results. A prospective study of men with extraprostatic disease randomized 82 patients to receive either leuprolide, goserelin, cyproterone acetate, or close clinical monitoring. All patients underwent serial cognitive assessments at baseline, at the start of treatment, and at 6 months.[32] A decrease in cognitive performance after 6 months of ADT use was found in the patients on active treatment compared with those on the close monitoring arm. A major limitation of this study is that spatial memory testing was omitted from the analysis, which has been shown to be more sensitive in detecting cognitive differences.[33] Another cross-sectional study compared neuropsychological parameters in three groups of patients: patients with prostate cancer receiving ADT, patients with prostate cancer not receiving ADT, and normal controls.[34] Here, the authors made a distinction between time-based prospective memory (eg, remembering to call a friend in 1 hour) and event-based prospective memory (eg, remembering to buy fruit when passing a fruit stand). This study demonstrated that men receiving ADT did demonstrably worse in event-related prospective memory than men not receiving ADT and matched controls. A limitation of this study was the lack of long-term follow-up on cognitive function, making it difficult to know whether the neurocognitive effects of ADT persist after discontinuation of therapy.
These studies can be contrasted with several others that have not demonstrated an association between use of ADT and neurocognitive decline. A large cohort study compared cognitive function using neuropsychological tests in the following groups: men with prostate cancer receiving continuous ADT, men with prostate cancer not receiving ADT, and healthy controls.[35] The authors found no difference in cognitive function over 12 months of ADT use. Furthermore, a recent large meta-analysis failed to find an association between cognitive impairment and ADT.[36] Similar findings were reported in one of the largest population studies of this kind, which examined 1.2 million Medicare beneficiaries who developed prostate cancer from 2001 to 2014.[37] The risk of dementia was not associated with duration of ADT (ie, there was no dose-dependent effect). Currently, there is mixed evidence on whether ADT is associated with long-term neurocognitive impairments, and this remains a topic of active research. In patients with baseline cognitive impairment, it is reasonable to attempt to minimize exposure to ADT, if possible (through intermittent treatment or by treating with anti-androgen monotherapy).
Thrombotic Complications
Thromboembolic risk has been noted in several studies.[31,38] In a large retrospective series, patients who received ADT had a 15% absolute risk of venous thromboembolism (VTE), compared with 7% among patients who did not receive ADT. Overall, there was a significant association between ADT use and risk of VTE or arterial embolism (adjusted hazard ratio [HR], 1.56; 95% CI, 1.50–1.61; P < .0001). Furthermore, greater duration of ADT was associated with an increased number of VTE events. These data are especially important to consider in patients with additional risk factors for VTE. Indeed, as indicated previously, tamoxifen has been reported to be useful therapy for ADT-associated gynecomastia,[13] yet it is an independent risk factor for thrombosis. Breast irradiation can be an alternative in symptomatic patients with thrombotic risk.
Data regarding use of ADT and risk of stroke are conflicting. A prospective case-control study from Taiwan reported no increased risk of stroke in patients with prostate cancer receiving ADT vs those not receiving it in an adjusted analysis.[39] In contrast, a meta-analysis reported a trend toward increased risk of stroke with ADT; however, this was not statistically significant (HR, 1.12; 95% CI, 0.95–1.32; P = .16). That said, subgroup analysis did identify GnRH agonist use alone or combined with anti-androgen use, and surgical castration, as having a significant association with stroke.[40] Based on the available evidence, risk of stroke should be mentioned when obtaining informed consent; however, the individual absolute risk is likely low.
Cardiovascular Complications
There are conflicting data on the risk of cardiovascular disease (CVD) with use of ADT. Currently, there are no prospective randomized controlled data assessing the association of ADT use with risk of CVD. In a large retrospective population-based cohort study, multivariate analyses demonstrated that men with newly diagnosed prostate cancer who received ADT had a 20% increased risk of CVD.[41] Somewhat paradoxically, the study found that men receiving treatment for less than 12 months were at higher risk for cardiovascular events compared with men who had been on ADT for longer. A major limitation of this study is that the researchers were unable to control for smoking and obesity, which are two major risk factors for CVD and could confound the results. Additionally, age was a major variable associated with CVD. A meta-analysis of eight large trials found no significant differences in risk of CVD in patients receiving ADT vs controls, nor was duration of ADT significantly associated with increased risk of CVD.[42]
KEY POINTS
·         Vasomotor complications are common in men receiving androgen deprivation therapy (ADT) and can be mitigated by pharmacologic and nonpharmacologic strategies.
·         There is an increased risk of osteoporosis and skeletal-related events in men receiving ADT. Calcium and vitamin D supplements should be offered to all patients. Osteoclast inhibitors should be considered in men at high risk for osteoporotic fractures and men with metastatic castration-resistant prostate cancer.
·         Evidence on the association between ADT use and neurocognitive decline and cardiovascular disease remains mixed and controversial.
·         Multidisciplinary management with specialists and allied health professionals may assist in managing the complex side effects of ADT.
Currently, no specific recommendation adjustments for men with prostate cancer receiving ADT should be pursued outside of primary prevention, including smoking cessation, and secondary prevention, including glycemic control, lipid-lowering therapy, and aspirin therapy for men with coronary artery disease. Given the propensity of patients on ADT to gain central adiposity, a healthy diet and exercise program are recommended to all patients at the start of treatment. Intermittent ADT or antiandrogen monotherapy may be appropriate for select patients (eg, those with biochemical relapse, low burden of metastases with more indolent course, and deep PSA nadir) at high risk for cardiovascular events; such patients should be evaluated on an individual basis and engaged in shared decision making, weighing the risk of a cardiovascular event vs cancer progression. Ultimately, patients with known CVD or those with numerous CVD risk factors should be evaluated promptly, if they develop new symptoms of chest pain or dyspnea while on ADT. A referral to cardio-oncology may assist in risk stratification.
Renal Complications
Observational data suggest an association between ADT use and rate of acute kidney injury (AKI).[43,44] In retrospective studies, the link appeared stronger with use of GnRH agonists plus an antiandrogen, as well as with GnRH agonists alone, compared with surgical castration; however, the inherent biases within such studies and the potential for numerous confounders raise questions regarding causality and the strength of association. Thus, use of ADT may present an increased risk of AKI, although the true risk and clinical significance are difficult to quantify.
Conclusion
ADT is the cornerstone of management for men with metastatic prostate cancer. It is associated with numerous side effects that can influence quality of life. Hot flushes, sexual dysfunction, gynecomastia, osteoporosis, metabolic syndrome, and depression are well-described complications of ADT. The implications of ADT use for risks of neurocognitive decline and CVD remain controversial, with multiple studies documenting conflicting results. Addressing quality-of-life concerns is of paramount importance with use of ADT, given the increased survival benefits found with combinations of ADT with other agents. Given the advanced age of many patients and the prolonged duration of ADT when treating localized disease with curative intent, some patients remain hypogonadal long after completion of therapy, underscoring the potential long-lasting sequelae of treatment. While consensus is lacking, there are mounting data[45] suggesting that testosterone replacement therapy may be cautiously used in select individuals in whom risk of prostate cancer relapse is determined to be low and hypogonadism remains long after ADT cessation. Ultimately, applying a multidisciplinary care model that incorporates specialists and allied healthcare professionals to aid in pharmacologic and lifestyle interventions is likely to provide optimal benefit in managing side effects for patients receiving long-term ADT.
Financial Disclosure: The authors have no significant financial interest in or other relationship with the manufacturer of any product or provider of any service mentioned in this article.
References:
1.       Sartor O, de Bono JS. Metastatic prostate cancer. N Engl J Med. 2018;378:645-57.
2.       Sharifi N, Gulley JL, Dahut WL. Androgen deprivation therapy for prostate cancer. JAMA. 2005;294:238-44.
3.       Holzbeierlein JM, McLaughlin MD, Thrasher JB. Complications of androgen deprivation therapy for prostate cancer. Curr Opin Urol. 2004;14:177-83.
4.       Smith JA Jr. Management of hot flushes due to endocrine therapy for prostate carcinoma. Oncology (Williston Park). 1996;10:1319-22; discussion 24.
5.       Irani J, Salomon L, Oba R, et al. Efficacy of venlafaxine, medroxyprogesterone acetate, and cyproterone acetate for the treatment of vasomotor hot flushes in men taking gonadotropin-releasing hormone analogues for prostate cancer: a double-blind, randomised trial. Lancet Oncol. 2010;11:147-54.
6.       Loprinzi CL, Dueck AC, Khoyratty BS, et al. A phase III randomized, double-blind, placebo-controlled trial of gabapentin in the management of hot flashes in men (N00CB). Ann Oncol. 2009;20:542-9.
7.       Shanafelt TD, Barton DL, Adjei AA, Loprinzi CL. Pathophysiology and treatment of hot flashes. Mayo Clin Proc. 2002;77:1207-18.
8.       Beer TM, Benavides M, Emmons SL, et al. Acupuncture for hot flashes in patients with prostate cancer. Urology. 2010;76:1182-8.
9.       Alemozaffar M, Regan MM, Cooperberg MR, et al. Prediction of erectile function following treatment for prostate cancer. JAMA. 2011;306:1205-14.
10.   Ferrer M, Suarez JF, Guedea F, et al. Health-related quality of life 2 years after treatment with radical prostatectomy, prostate brachytherapy, or external beam radiotherapy in patients with clinically localized prostate cancer. Int J Radiat Oncol Biol Phys. 2008;72:421-32.
11.   Gay HA, Sanda MG, Liu J, et al. External beam radiation therapy or brachytherapy with or without short-course neoadjuvant androgen deprivation therapy: results of a multicenter, prospective study of quality of life. Int J Radiat Oncol Biol Phys. 2017;98:304-17.
12.   Tunio MA, Al-Asiri M, Al-Amro A, et al. Optimal prophylactic and definitive therapy for bicalutamide-induced gynecomastia: results of a meta-analysis. Curr Oncol. 2012;19:e280-e288.
13.   Viani GA, Bernardes da Silva LG, Stefano EJ. Prevention of gynecomastia and breast pain caused by androgen deprivation therapy in prostate cancer: tamoxifen or radiotherapy? Int J Radiat Oncol Biol Phys. 2012;83:e519-e524.
14.   Higano CS. Bone loss and the evolving role of bisphosphonate therapy in prostate cancer. Urol Oncol. 2003;21:392-8.
15.   Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of fracture after androgen deprivation for prostate cancer. N Engl J Med. 2005;352:154-64.
16.   Smith MR, McGovern FJ, Zietman AL, et al. Pamidronate to prevent bone loss during androgen-deprivation therapy for prostate cancer. N Engl J Med. 2001;345:948-55.
17.   Saad F, Gleason DM, Murray R, et al. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst. 2002;94:1458-68.
18.   Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet Oncol. 2011;377:813-22.
19.   Smith MR, Halabi S, Ryan CJ, et al. Randomized controlled trial of early zoledronic acid in men with castration-sensitive prostate cancer and bone metastases: results of CALGB 90202 (Alliance). J Clin Oncol. 2014;32:1143-50.
20.   Braunstein LZ, Chen MH, Loffredo M, et al. Obesity and the odds of weight gain following androgen deprivation therapy for prostate cancer. Prostate Cancer. 2014;2014:230812.
21.   Kim HS, Moreira DM, Smith MR, et al. A natural history of weight change in men with prostate cancer on androgen-deprivation therapy (ADT): results from the Shared Equal Access Regional Cancer Hospital (SEARCH) database. BJU Int. 2011;107:924-8.
22.   Seible DM, Gu X, Hyatt AS, et al. Weight gain on androgen deprivation therapy: which patients are at highest risk? Urology. 2014;83:1316-21.
23.   Mitsuzuka K, Kyan A, Sato T, et al. Influence of 1 year of androgen deprivation therapy on lipid and glucose metabolism and fat accumulation in Japanese patients with prostate cancer. Prostate Cancer Prostatic Dis. 2016;19:57-62.
24.   Salvador C, Planas J, Agreda F, et al. Analysis of the lipid profile and atherogenic risk during androgen deprivation therapy in prostate cancer patients. Urol Int. 2013;90:41-4.
25.   Torimoto K, Samma S, Kagebayashi Y, et al. The effects of androgen deprivation therapy on lipid metabolism and body composition in Japanese patients with prostate cancer. Jpn J Clin Oncol. 2011;41:577-81.
26.   Bosco C, Crawley D, Adolfsson J, et al. Quantifying the evidence for the risk of metabolic syndrome and its components following androgen deprivation therapy for prostate cancer: a meta-analysis. PLoS One. 2015;10:e0117344.
27.   Braga-Basaria M, Dobs AS, Muller DC, et al. Metabolic syndrome in men with prostate cancer undergoing long-term androgen-deprivation therapy. J Clin Oncol. 2006;24:3979-83.
28.   Chang D, Joseph DJ, Ebert MA, et al. Effect of androgen deprivation therapy on muscle attenuation in men with prostate cancer. J Med Imaging Radiat Oncol. 2014;58:223-8.
29.   Winters-Stone KM, Moe E, Graff JN, et al. Falls and frailty in prostate cancer survivors: current, past, and never users of androgen deprivation therapy. J Am Geriatr Soc. 2017;65:1414-9.
30.   Shahinian VB, Kuo YF, Freeman JL, Goodwin JS. Risk of the ‘androgen deprivation syndrome’ in men receiving androgen deprivation for prostate cancer. Arch Intern Med. 2006;166:465-71.
31.   Nead KT, Gaskin G, Chester C, et al. Association between androgen deprivation therapy and risk of dementia. JAMA Oncol. 2017;3:49-55.
32.   Green HJ, Pakenham KI, Headley BC, et al. Altered cognitive function in men treated for prostate cancer with luteinizing hormone-releasing hormone analogues and cyproterone acetate: a randomized controlled trial. BJU Int. 2002;90:427-32.
33.   Cherrier MM, Asthana S, Plymate S, et al. Testosterone supplementation improves spatial and verbal memory in healthy older men. Neurology. 2001;57:80-8.
34.   Yang J, Zhong F, Qiu J, et al. Dissociation of event-based prospective memory and time-based prospective memory in patients with prostate cancer receiving androgen-deprivation therapy: a neuropsychological study. Eur J Cancer Care (Engl). 2015;24:198-204.
35.   Alibhai SM, Breunis H, Timilshina N, et al. Impact of androgen-deprivation therapy on cognitive function in men with nonmetastatic prostate cancer. J Clin Oncol. 2010;28:5030-7.
36.   Sun M, Cole AP, Hanna N, et al. Cognitive impairment in men with prostate cancer treated with androgen deprivation therapy: a systematic review and meta-analysis. J Urol. 2018;199:1417-25.
37.   Baik SH, Kury FSP, McDonald CJ. Risk of Alzheimer’s disease among senior Medicare beneficiaries treated with androgen deprivation therapy for prostate cancer. J Clin Oncol. 2017;35:3401-9.
38.   Ehdaie B, Atoria CL, Gupta A, et al. Androgen deprivation and thromboembolic events in men with prostate cancer. Cancer. 2012;118:3397-406.
39.   Chung SD, Chen YK, Wu FJ, Lin HC. Hormone therapy for prostate cancer and the risk of stroke: a 5-year follow-up study. BJU Int. 2012;109:1001-5.
40.   Meng F, Zhu S, Zhao J, et al. Stroke related to androgen deprivation therapy for prostate cancer: a meta-analysis and systematic review. BMC Cancer. 2016;16:180.
41.   Saigal CS, Gore JL, Krupski TL, et al. Androgen deprivation therapy increases cardiovascular morbidity in men with prostate cancer. Cancer. 2007;110:1493-500.
42.   Nguyen PL, Alibhai SM, Basaria S, et al. Adverse effects of androgen deprivation therapy and strategies to mitigate them. Eur Urol. 2015;67:825-36.
43.   Gandaglia G, Sun M, Hu JC, et al. Gonadotropin-releasing hormone agonists and acute kidney injury in patients with prostate cancer. Eur Urol. 2014;66:1125-32.
44.   Lapi F, Azoulay L, Niazi MT, et al. Androgen deprivation therapy and risk of acute kidney injury in patients with prostate cancer. JAMA. 2013;310:289-96.
45.   Golla V, Kaplan AL. Testosterone therapy on active surveillance and following definitive treatment for prostate cancer. Curr Urol Rep. 2017;18:49

--------------------------------------------------------------------------------------------------------------------------------------

Aggressive prostate and lung cancers are driven by common mechanisms, researchers find

Date:  October 4, 2018     Source:    University of California - Los Angeles Health Sciences
Summary:
    Researchers have discovered a common process in the development of late-stage, small cell cancers of the prostate and lung. These shared molecular mechanisms could lead to the development of drugs to treat not just prostate and lung cancers, but small cell cancers of almost any organ.
https://www.sciencedaily.com/images/2018/10/181004155432_1_540x360.jpg
A microscopic image of small cell neuroendocrine prostate cancer, with cancer cells expressing diagnostic prostate cancer markers in green and red. Blue areas indicate cell nuclei.
Credit: Jung Wook Park and Owen Witte
UCLA researchers have discovered a common process in the development of late-stage, small cell cancers of the prostate and lung. These shared molecular mechanisms could lead to the development of drugs to treat not just prostate and lung cancers, but small cell cancers of almost any organ.
The key finding: Prostate and lung cells have very different patterns of gene expression when they're healthy, but almost identical patterns when they transform into small cell cancers. The research suggests that different types of small cell tumors evolve similarly, even when they come from different organs.
The study, led by Dr. Owen Witte, founding director of the UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research and professor of microbiology, immunology and molecular genetics, was published in the journal Science. Witte collaborated with scientists from UCLA's Crump Institute for Molecular Imaging and the UCLA Jonsson Comprehensive Cancer Center.
Cancers that become resistant to treatment often develop into small cell cancers -- also known as small cell neuroendocrine carcinomas, or SCNCs -- which generally have extremely poor prognoses. Certain cancers can evade treatment in part by changing cell types -- from aggressive adenocarcinoma to small cell carcinoma, for example.
Previous research hinted that small cell cancers from different organs may be driven by common mechanisms, but the UCLA study is the first to so clearly describe the steps in their evolution.
"Small cell cancers of the lung, prostate, bladder, and other tissues were long thought to be similar in name alone -- and they were treated by oncologists as different entities," Witte said. "Over the past few years, though, researchers have increasingly begun to realize that there are similarities in the cancers, and that's what our work confirms."
Dr. Jung Wook Park, the study's first author, and UCLA collaborators explored the potential parallels between the cancer types by transplanting human prostate cells with five genes, known collectively as PARCB, into mice. When those cells grew in the mice, they displayed unique features of human small cell neuroendocrine carcinomas.
The team also identified that for small cell neuroendocrine carcinomas to develop in the prostate, two tumor suppressor genes, TP53 and RB1, which are known for protecting normal cells from transforming into cancer cells, had to be simultaneously inactivated when PARCB was introduced.
Additional tests confirmed striking similarities between the PARCB-SCNC cells and small cell prostate cancer cells from humans. In particular, RNA expression and the turning on and off of certain genes were nearly identical.
"The similarities between the PARCB-SCNC cancers and human small cell prostate cancer samples were extraordinary," Witte said. "If you blindly gave the data sets to any statistician, they would think they were the same cells."
The team also looked at large databases of gene expression, to compare the patterns of gene expression in their PARCB-SCNC cells to cancers of other organs. They found that the pattern of gene expression in PARCB-SCNC cells was extremely similar to those of both prostate and lung small cell cancers.
Next, they tested whether PARCB genes could alter healthy cells from human lungs into small cell lung cancers, and the scientists found that they could.
The team now is working on mapping which genes control the entire cascade of events that underlies the transition to small cell cancer.
"Our study revealed shared 'master gene regulators' -- the key proteins that control expression of multiple genes in small cell cancer cells," Witte said. "Studying the network of the master gene regulators could lead to a new way of combating deadly cancers."
The research was supported by the Broad Stem Cell Research Center Stem Cell Training Program and Hal Gaba Fund for Prostate Cancer Research, the UCLA Medical Scientist Training Program, the UCLA Specialized Program of Research Excellence in Prostate Cancer, the National Institutes of Health, the National Cancer Institute, the Prostate Cancer Foundation, the Department of Defense, the American Cancer Society and the W.M. Keck Foundation.
Story Source:
Materials provided by University of California - Los Angeles Health Sciences. Original written by Alice Walton. Note: Content may be edited for style and length.
Journal Reference:
    Jung Wook Park, John K. Lee, Katherine M. Sheu, Liang Wang, Nikolas G. Balanis, Kim Nguyen, Bryan A. Smith, Chen Cheng, Brandon L. Tsai, Donghui Cheng, Jiaoti Huang, Siavash K. Kurdistani, Thomas G. Graeber, Owen N. Witte. Reprogramming normal human epithelial tissues to a common, lethal neuroendocrine cancer lineage. Science, 2018; 362 (6410): 91 DOI: 10.1126/science.aat5749
 


Additional Links:

Prostatepedia Weekly 10.4.18

Aggressive prostate and lung cancers are driven by common mechanisms, researchers find -- ScienceDaily: Researchers have discovered a common process in the development of late-stage, small cell cancers of the prostate and lung. These shared molecular mechanisms could lead to the development of drugs to treat not just prostate and lung cancers, but small cell cancers of almost any organ.

Can aspirin help treat cancer?: A systematic review of 71 studies suggests that regular aspirin intake may help treat cancers such as colon cancer, breast cancer, and prostate cancer.

Prostate cancer outcomes today vs. outcomes yesteryear | THE "NEW" PROSTATE CANCER INFOLINK: As some of us have long suspected and understood, using data from men treated 20+ years ago as a guide to what would happen if you were treated the same way today is not really a very good idea. A …

Multimodal Therapy May Up Survival in Advanced Prostate Cancer: But the notable survival advantage seen with primary treatment consisting of radical prostatectomy plus radiotherapy also came with a high cost to quality of life.

Free randomized clinical trial of gallium-68-PSMA-11 PET indicator | THE "NEW" PROSTATE CANCER INFOLINK: A randomized clinical trial of the 68Ga-PSMA-11 PET indicator for men with a recurrence after radical prostatectomy has just been initiated at the University of California Los Angeles (UCLA). Eligi…

68Ga-PSMA-11 PET/CT in Newly Diagnosed Prostate Adenocarcino... : Clinical Nuclear Medicine: newly diagnosed PCa.
Materials and Methods
A total of 78 patients with biopsy-proven PCa who were referred for 68Ga-PSMA-11 PET/CT for primary staging were retrospectively analyzed. The patients were divided into risk groups according to the D'Amico risk stratification criteria. All of the patients had undergone pelvic MRI, and 65 patients had bone scintigraphy also. The findings of 68Ga-PSMA-11 PET/CT were compared with these conventional imaging (CI) methods for staging of the disease. The relations between SUVmax of the primary tumors and Gleason scores (GSs), prostate-specific antigen (PSA) levels, and metastatic extent of the disease were analyzed.
Results
Of 78 patients, 5 patients were in low-risk group, 18 patients were in intermediate-risk group, and 55 patients were in high-risk group. Metastatic disease was found in 40 (51.2%) of 78 patients in 68Ga-PSMA-11 PET/CT. Ten patients had regional lymph node metastases, and 30 patients had distant metastases. 68Ga-PSMA-11 PET/CT changed the staging in 44 (56.4%) of 78 patients compared with CI. There was significant difference between the SUVmax of the tumors with GSs of 6 and 7 compared with GSs of 8, 9, and 10 (P = 0.003).

Radical Prostatectomy Plus Radiotherapy Extends OS in Prostate Cancer | Cancer Network: A study in Cancer found a lower risk of prostate cancer–specific death and improved overall survival when patients were treated with this combination.

[Concordance evaluation of gleason score between laparoscopic prostatectomy and needle biopsy specimens]. - PubMed - NCBI: Wiad Lek. 2018;71(6):1193-1199. English Abstract

Salvage extended field or involved field nodal irradiation in 18F-fluorocholine PET/CT oligorecurrent nodal failures from prostate cancer | SpringerLink: Purpose The concept of metastasis-directed therapy for nodal oligorecurrences with stereotactic body radiotherapy is increasingly accepted. Hence, the comparison between salvage extended field...

No comments: