Physiology and Endocrinology of Hot Flashes in Prostate Cancer

2007 ◽  
Vol 1 (1) ◽  
pp. 8-17 ◽  
Author(s):  
Christine A. Engstrom ◽  
Christine E. Kasper

The purpose of this article is to integrate the physiology of the male reproductive system and the role of hormones in the pathophysiology and treatment of prostate cancer. The primary focus is to review hormonal changes associated with androgen ablation treatment and to integrate the available hormonal data into a hypothesis. This review used a systematic search of Medline references from 1990 to 2006. All sources were critically evaluated to arrive at an understanding of androgen deprivation symptoms, such as hot flushes/flashes, and to identify research needed in this area. Research is needed to explore the physiological mechanisms of hot flashes to develop better therapeutic treatment options to ameliorate side effects of hormonal treatment. Studies are needed to investigate all aspects of hot flashes in populations other than those with breast cancer, such as men with prostate cancer, carcinoid tumors, medullary thyroid tumors, pancreatic islet-cell tumors, renal cell carcinoma, and phenochromocytoma.

2020 ◽  
Vol 19 (1) ◽  
pp. 15-20
Author(s):  
Junyi Xiang ◽  
Feng Huang ◽  
Renhua Huang ◽  
Jingzhan Su ◽  
Yulong Liu

Prostate cancer is one of the leading causes of death in men all over the world. Treatment options such as androgen ablation therapy and cytotoxic agents have many undesirable side effects, narrow therapeutic windows, or other limitations. In this research, we have explored the effects of paeonol on prostate cancer and its mechanism of action. Our results have shown that paeonol reduced the viability of prostate cancer cells in a dose-dependent manner. The wound-healing assay, a surrogate marker of tumor metastasis, showed that the relative wound width of 10 µM group was less than that of 50 µM paeonol-treated cells. Besides, the results of the transwell assay also showed that the number of migrated cells was significantly lower after treatment with 50 µM paeonol compared to the 10 µM group. The Western blot results showed that paeonol treatment induced a decrease in the mesenchymal markers (vimentin and N-cadherin), while the epithelial marker (E-cadherin) increased in a dose-dependent manner suggesting that paeonol effectively inhibits the epithelial-mesenchymal transformation in PC3 cells. Furthermore, the expression of STAT3 and p-STAT3 was also decreased after paeonol treatment, which indicated that the STAT3 signaling pathway was inhibited by paeonol. To conclude, the results summarized in this paper suggest that paeonol could be a potential candidate in the treatment of prostate cancer.


Author(s):  
Stephan H. Flüchter ◽  
Ralf Weiser ◽  
Christoph Gamper

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Joanna Cyrta ◽  
Anke Augspach ◽  
Maria Rosaria De Filippo ◽  
Davide Prandi ◽  
Phillip Thienger ◽  
...  

Abstract Advanced prostate cancer initially responds to hormonal treatment, but ultimately becomes resistant and requires more potent therapies. One mechanism of resistance observed in around 10–20% of these patients is lineage plasticity, which manifests in a partial or complete small cell or neuroendocrine prostate cancer (NEPC) phenotype. Here, we investigate the role of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling complex in NEPC. Using large patient datasets, patient-derived organoids and cancer cell lines, we identify mSWI/SNF subunits that are deregulated in NEPC and demonstrate that SMARCA4 (BRG1) overexpression is associated with aggressive disease. We also show that SWI/SNF complexes interact with different lineage-specific factors in NEPC compared to prostate adenocarcinoma. These data point to a role for mSWI/SNF complexes in therapy-related lineage plasticity, which may also be relevant for other solid tumors.


Author(s):  
Pervin Hurmuz ◽  
Fadıl Akyol ◽  
Melis Gultekin ◽  
Gozde Yazici ◽  
Sezin Yuce Sari ◽  
...  

2016 ◽  
Vol 69 (5) ◽  
pp. 802-820 ◽  
Author(s):  
Roderick C.N. van den Bergh ◽  
Niels J. van Casteren ◽  
Thomas van den Broeck ◽  
Eve R. Fordyce ◽  
William K.M. Gietzmann ◽  
...  

2016 ◽  
Vol 9 (1) ◽  
pp. 105-105
Author(s):  
L. Kurtz Almog ◽  

Objective: According to the statistics, one in six men develops prostate cancer. There are several therapeutic options for prostate cancer. Anti-hormone therapy is one of treatment leading to an 80–90% remission. A significant percentage of men who received anti-hormonal treatment complain of decreased libido and erectile dysfunction. Similarly, these men may also suffer from hot flashes, weight gain, growth of [male] breast tissue (gynecomastia), lack of energy and initiative, depression and mental disorders. In this paper, I would like to present a therapeutic case of a man who received sexual counseling at the Israel Cancer Association, and who, upon receiving antihormonal treatment, changed his sexual orientation after having contracted prostate cancer. Design and Method: A series of meetings were conducted, encompassing an interview and sexual counseling. The Klein Sexual Orientation Grid (KSOG) was used in order to evaluate the patient’s sexual orientation. Results: According to the GRID several parameters such as sexual attraction and sexual behavior were significantly different in the comparison between the past and the present. Parameters such as social preference and hetero/gay style did not change. The remaining results will be presented. Conclusions: Following an analysis of this case, it may be concluded that a person’s sexual orientation can change during his lifetime. The hypothesis is that a life-changing event such as cancer, including all the side effects of difficult antihormonal treatments, has the potential to evoke a change in the person’s overall experience and in his sexual experience in particular.


2020 ◽  
Vol 38 (6_suppl) ◽  
pp. 235-235
Author(s):  
Robert H. Getzenberg ◽  
Mark C. Scholz ◽  
Alexandra Scholz ◽  
Mitchell S. Steiner

235 Background: Androgen Deprivation Therapy (ADT) is a mainstay in the treatment of advanced prostate cancer. ADT-induced estrogen deficiency related side effects may cause men to delay, pause, or discontinue ADT, increases morbidity and mortality, and can significantly impact quality of life. ADT-induced effects include hot flashes, bone loss and fractures, fatigue, decreased libido, and metabolic and lipid changes. Currently there are no FDA approved treatments for ADT-induced hot flashes in men with advanced prostate cancer. In this study, a survey was conducted on the impact of hot flashes, one of the hallmark ADT-induced estrogen deficiency effects, in a contemporary cohort. Methods: During the period of August/September 2019, 212 men with advanced prostate cancer on ADT participated in a digital survey conducted by the Prostate Cancer Research Institute (PCRI) focused on the frequency, severity and impact of their hot flashes. The men were at least 50 years of age with 61% being 70 or older. ADT types included LUPRONÒ(64%), ELIGARDÒ(12%), ZOLADEXÒ(7%) and other forms of hormonal therapy (17%). Results: Of the 212 men surveyed, 99% reported hot flashes with 80% indicating that they experience clinically significant, moderate to severe hot flashes. 77% of men reported that the number of hot flashes stayed the same or increased during their hormonal therapy. 37% of the men experienced more than 5 hot flashes per day and 23% indicated that they felt embarrassed about their hot flashes. Only 51% had either researched how to address their hot flashes or discussed them with their physician. Importantly, 16% considered halting ADT as a result of their hot flashes. Conclusions: This contemporary survey underscores the significant unmet medical need to treat moderate to severe hot flashes which occurred in 80% of the men studied. As about half of the men have not discussed their symptoms with a physician either because of embarrassment or lack of treatment options, the number of men with moderate to severe hot flashes appears to be greatly under-reported. As men on ADT are living longer with prostate cancer, finding an effective and safe treatment for debilitating hot flashes must be a priority.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS5087-TPS5087
Author(s):  
Kim N. Chi ◽  
Ur Metser ◽  
Johannes Czernin ◽  
Jeremie Calais ◽  
Vikas Prasad ◽  
...  

TPS5087 Background: Treatment options with minimal toxicity and novel mechanisms of action are urgently needed to improve clinical outcomes from mCRPC. Prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) represents a new treatment for patients with PSMA-avid mCRPC. 177Lu-PNT2002 (also known as [Lu-177]-PSMA-I&T) is a PSMA-targeting agent and studies have shown demonstrable promising initial data. This trial seeks to prospectively evaluate the efficacy of 177Lu-PNT2002 for men with progressive mCRPC after androgen receptor axis-targeted (ARAT) therapy. Methods: This is a multi-center, open-label, phase III study. All patients must be at least 18 years of age, have documented progressive mCRPC at time of screening, high PSMA expression by PSMA PET/CT per blinded independent central review (BICR), chemotherapy naïve for CRPC and unfit or unwilling to receive chemotherapy. The study will commence with a 25-patient dosimetry lead-in. In the dosimetry phase, patients will receive up to four cycles of 177Lu-PNT2002 at 6.8 GBq every 8 weeks. In the randomization phase, approximately 390 patients will be randomized in a 2:1 ratio to receive 177Lu-PNT2002 (Arm A) versus enzalutamide or abiraterone (with prednisone or dexamethasone) (Arm B). Patients randomized to Arm B have an option to crossover to 177Lu-PNT2002 treatment after BICR-assessed radiologic progression. The primary endpoint is Radiological progression-free survival (rPFS) assessed by BICR using Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 (soft tissue) and Prostate Cancer Working Group 3 (PCWG3) (bone) criteria. Key secondary endpoints include objective response rate, duration of response, PSA response, and overall survival. The study is powered at 90% to test the alternative hypothesis of a hazard ratio (HR) ≤ 0.66 at an α of 0.025. ClinicalTrials.gov identifier: NCT04647526. Clinical trial information: NCT04647526.


At JADPRO Live Virtual 2020, Brenda Martone, MSN, ANP-BC, AOCNP®, discussed treatment options available for advanced prostate cancer patients and the role of genetics and genomics in patients with advanced prostate cancer.


Author(s):  
Leslie C. Costello ◽  
Renty B. Franklin

Hormone-independent malignancy is a major issue of morbidity and deaths that confronts prostate cancer. Despite decades of research, the oncogenic and hormonal implications in the development and progression of prostate malignancy remain mostly speculative. This is largely due to the absence and/or lack of consideration by contemporary clinicians and biomedical investigators regarding the established implications of the co-regulation of testosterone and prolactin in the development, maintenance, metabolism and functions of the prostate gland. Especially relevant is the major metabolic function of production of high levels of citrate by the peripheral zone acinar epithelial cells. Citrate production, along with growth and proliferation by these cells, is regulated by co-existing testosterone and prolactin signaling pathways; and by the oncogenic down-regulation of ZIP1 transporter/zinc/citrate in the development of malignancy. These relationships had not been considered in the issues of hormonedependent malignancy. This review provides the relevant background that has established the dual role of testosterone and prolactin regulation of the prostate gland; which is essential to address the implications in the oncogenic development and progression of hormone-dependent malignancy. The oncogenic factor along with testosterone-dependent and prolactin-dependent relationships leads to the plausible concept that androgen ablation for the treatment of testosterone-dependent malignancy results in the development of prolactin-dependent malignancy; which is testosterone-independent malignancy. Consequently, both testosterone ablation and prolactin ablation are required to prevent and/or abort terminal hormonedependent prostate cancer.


Sign in / Sign up

Export Citation Format

Share Document