Siegel, R. L., Miller, Okay. D., Wagle, N. S. & Jemal, A. Most cancers statistics, 2023. CA Most cancers J. Clin. 73, 17–48 (2023).
Ma, B., Wells, A., Wei, L. & Zheng, J. Prostate most cancers liver metastasis: dormancy and resistance to remedy. Semin. Most cancers Biol. 71, 2–9 (2021).
Tsilimigras, D. I. et al. Liver metastases. Nat. Rev. Dis. Prim. 7, 27 (2021).
Gandaglia, G. et al. Affect of the location of metastases on survival in sufferers with metastatic prostate most cancers. Eur. Urol. 68, 325–334 (2015).
Pond, G. R. et al. The prognostic significance of metastatic web site in males with metastatic castration-resistant prostate most cancers. Eur. Urol. 65, 3–6 (2014).
Halabi, S. et al. Meta-analysis evaluating the influence of web site of metastasis on general survival in males with castration-resistant prostate most cancers. J. Clin. Oncol. 34, 1652–1659 (2016).
Heck, M. M. et al. Remedy end result, toxicity, and predictive elements for radioligand remedy with (177)Lu-PSMA-I&T in metastatic castration-resistant prostate most cancers. Eur. Urol. 75, 920–926 (2019).
Rahbar, Okay. et al. PSMA focused radioligand remedy in metastatic castration resistant prostate most cancers after chemotherapy, abiraterone and/or enzalutamide. A retrospective evaluation of general survival. Eur. J. Nucl. Med. Mol. Imaging 45, 12–19 (2018).
Beltran, H. et al. A section II trial of the Aurora kinase A inhibitor alisertib for sufferers with castration-resistant and neuroendocrine prostate most cancers: efficacy and biomarkers. Clin. Most cancers Res. 25, 43–51 (2019).
Gandaglia, G. et al. Distribution of metastatic websites in sufferers with prostate most cancers: a population-based evaluation. Prostate 74, 210–216 (2014).
Haffner, M. C. et al. Monitoring the clonal origin of deadly prostate most cancers. J. Clin. Make investments. 123, 4918–4922 (2013).
van Dessel, L. F. et al. Utility of circulating tumor DNA in potential scientific oncology trials — standardization of preanalytical situations. Mol. Oncol. 11, 295–304 (2017).
von Eyben, F. E., Picchio, M., von Eyben, R., Rhee, H. & Bauman, G. 68Ga-labeled prostate-specific membrane antigen ligand positron emission tomography/computed tomography for prostate most cancers: a scientific evaluation and meta-analysis. Eur. Urol. Focus. 4, 686–693 (2018).
Damjanovic, J. et al. 68Ga-PSMA-PET/CT for the analysis of liver metastases in sufferers with prostate most cancers. Most cancers Imaging 19, 37 (2019).
Roviello, G., Petrioli, R., Villari, D. & D’Angelo, A. Treating de novo metastatic castration-sensitive prostate most cancers with visceral metastases: an evolving subject. Clin. Genitourin. Most cancers 19, 83–86 (2021).
Khreish, F. et al. Response and end result of liver metastases in sufferers with metastatic castration-resistant prostate most cancers (mCRPC) present process 177Lu-PSMA-617 radioligand remedy. Eur. J. Nucl. Med. Mol. Imaging 48, 103–112 (2021).
Bubendorf, L. et al. Metastatic patterns of prostate most cancers: an post-mortem examine of 1,589 sufferers. Hum. Pathol. 31, 578–583 (2000).
Shah, R. B. et al. Androgen-independent prostate most cancers is a heterogeneous group of ailments: classes from a speedy post-mortem program. Most cancers Res. 64, 9209–9216 (2004).
Pezaro, C. et al. Visceral illness in castration-resistant prostate most cancers. Eur. Urol. 65, 270–273 (2014).
van den Bergh, G. P. A. et al. Incidence and survival of castration-resistant prostate most cancers sufferers with visceral metastases: outcomes from the Dutch CAPRI-registry. Prostate Most cancers Prostatic Dis. 26, 162–169 (2023).
Deng, Y. et al. A surveillance, epidemiology and finish outcomes database evaluation of the prognostic worth of organ-specific metastases in sufferers with superior prostatic adenocarcinoma. Oncol. Lett. 18, 1057–1070 (2019).
Shou, J., Zhang, Q., Wang, S. & Zhang, D. The prognosis of various distant metastases sample in prostate most cancers: a inhabitants based mostly retrospective examine. Prostate 78, 491–497 (2018).
Cotogno, P. M., Ranasinghe, L. Okay., Ledet, E. M., Lewis, B. E. & Sartor, O. Laboratory-based biomarkers and liver metastases in metastatic castration-resistant prostate most cancers. Oncologist 23, 791–797 (2018).
Ranasinghe, L. et al. Relationship between serum markers and quantity of liver metastases in castration-resistant prostate most cancers. Most cancers Deal with. Res. Commun. 20, 100151 (2019).
Rahbar, Okay. et al. German multicenter examine investigating 177Lu-PSMA-617 radioligand remedy in superior prostate most cancers sufferers. J. Nucl. Med. 58, 85–90 (2017).
Ahmadzadehfar, H. et al. Prior therapies as prognostic elements of general survival in metastatic castration-resistant prostate most cancers sufferers handled with [177Lu]Lu-PSMA-617. A WARMTH multicenter examine (the 617 trial). Eur. J. Nucl. Med. Mol. Imaging 48, 113–122 (2021).
Kessel, Okay. et al. Second line chemotherapy and visceral metastases are related to poor survival in sufferers with mCRPC receiving 177Lu-PSMA-617. Theranostics 9, 4841–4848 (2019).
Luna-Gutierrez, M. et al. Bettering general survival and high quality of life in sufferers with prostate most cancers and neuroendocrine tumors utilizing 177Lu-iPSMA and 177Lu-DOTATOC: expertise after 905 therapy doses. Pharmaceutics 15, 1988 (2023).
Fendler, W. P. et al. Prostate-specific membrane antigen ligand positron emission tomography in males with nonmetastatic castration-resistant prostate most cancers. Clin. Most cancers Res. 25, 7448–7454 (2019).
Hofman, M. S. et al. Prostate-specific membrane antigen PET-CT in sufferers with high-risk prostate most cancers earlier than curative-intent surgical procedure or radiotherapy (proPSMA): a potential, randomised, multicentre examine. Lancet 395, 1208–1216 (2020).
Maitland, N. J. Resistance to antiandrogens in prostate most cancers: is it inevitable, intrinsic or induced? Cancers 13, 327 (2021).
Buxton, A. Okay., Abbasova, S., Bevan, C. L. & Leach, D. A. Liver microenvironment response to prostate most cancers metastasis and hormonal remedy. Cancers 14, 6189 (2022).
Berry, W. R., Laszlo, J., Cox, E., Walker, A. & Paulson, D. Prognostic elements in metastatic and hormonally unresponsive carcinoma of the prostate. Most cancers 44, 763–775 (1979).
Elements within the prognosis of carcinoma of the prostate: a cooperative examine. The Veterans Administration Cooperative Urological Analysis Group. J. Urol. 100, 59–65 (1968).
Petrylak, D. P., Scher, H. I., Li, Z., Myers, C. E. & Geller, N. L. Prognostic elements for survival of sufferers with bidimensionally measurable metastatic hormone-refractory prostatic most cancers handled with single-agent chemotherapy. Most cancers 70, 2870–2878 (1992).
Chi, Okay. N. et al. A prognostic index mannequin for predicting general survival in sufferers with metastatic castration-resistant prostate most cancers handled with abiraterone acetate after docetaxel. Ann. Oncol. 27, 454–460 (2016).
Conteduca, V. et al. Affect of visceral metastases on end result to abiraterone after docetaxel in castration-resistant prostate most cancers sufferers. Future Oncol. 11, 2881–2891 (2015).
Vasil’eva, N. N. & Milievskaia, I. L. [Neurogenic stomach neoplasms induced by nitrosomethylures in Syrian hamsters]. Arkh Patol. 39, 66–71 (1977).
Armstrong, A. J. et al. A up to date prognostic nomogram for males with hormone-refractory metastatic prostate most cancers: a TAX327 examine evaluation. Clin. Most cancers Res. 13, 6396–6403 (2007).
Loriot, Y. et al. Enzalutamide in castration-resistant prostate most cancers sufferers with visceral illness within the liver and/or lung: outcomes from the randomized managed section 3 AFFIRM trial. Most cancers 123, 253–262 (2017).
Alumkal, J. J. et al. Impact of visceral illness web site on outcomes in sufferers with metastatic castration-resistant prostate most cancers handled with enzalutamide within the PREVAIL trial. Clin. Genitourin. Most cancers 15, 610–617 e613 (2017).
Beer, T. M. et al. Enzalutamide in metastatic prostate most cancers earlier than chemotherapy. N. Engl. J. Med. 371, 424–433 (2014).
Baciarello, G. et al. Affect of abiraterone acetate plus prednisone in sufferers with castration-sensitive prostate most cancers and visceral metastases over 4 years of follow-up: a post-hoc exploratory evaluation of the LATITUDE examine. Eur. J. Most cancers 162, 56–64 (2022).
Satapathy, S., Mittal, B. R. & Sood, A. Visceral metastases as predictors of response and survival outcomes in sufferers of castration-resistant prostate most cancers handled with 177Lu-labeled prostate-specific membrane antigen radioligand remedy: a scientific evaluation and meta-analysis. Clin. Nucl. Med. 45, 935–942 (2020).
Armstrong, A. J. et al. Improvement and validation of a prognostic mannequin for general survival in chemotherapy-naive males with metastatic castration-resistant prostate most cancers. Ann. Oncol. 29, 2200–2207 (2018).
Akamatsu, S. et al. Improvement and validation of a novel prognostic mannequin for predicting general survival in treatment-naive castration-sensitive metastatic prostate most cancers. Eur. Urol. Oncol. 2, 320–328 (2019).
Gafita, A. et al. Nomograms to foretell outcomes after 177Lu-PSMA remedy in males with metastatic castration-resistant prostate most cancers: a global, multicentre, retrospective examine. Lancet Oncol. 22, 1115–1125 (2021).
Labrecque, M. P. et al. Molecular profiling stratifies various phenotypes of treatment-refractory metastatic castration-resistant prostate most cancers. J. Clin. Make investments. 129, 4492–4505 (2019).
Aggarwal, R. et al. Scientific and genomic characterization of treatment-emergent small-cell neuroendocrine prostate most cancers: a multi-institutional potential examine. J. Clin. Oncol. 36, 2492–2503 (2018).
Bakht, M. Okay. et al. Panorama of prostate-specific membrane antigen heterogeneity and regulation in AR-positive and AR-negative metastatic prostate most cancers. Nat. Most cancers 4, 699–715 (2023).
Feng, E. et al. Intrinsic molecular subtypes of metastatic castration-resistant prostate most cancers. Clin. Most cancers Res. 28, 5396–5404 (2022).
Conteduca, V. et al. Scientific options of neuroendocrine prostate most cancers. Eur. J. Most cancers 121, 7–18 (2019).
Thysell, E. et al. Gene expression profiles outline molecular subtypes of prostate most cancers bone metastases with totally different outcomes and morphology traceable again to the first tumor. Mol. Oncol. 13, 1763–1777 (2019).
Ku, S. Y., Gleave, M. E. & Beltran, H. In the direction of precision oncology in superior prostate most cancers. Nat. Rev. Urol. 16, 645–654 (2019).
Paschalis, A. et al. Prostate-specific membrane antigen heterogeneity and DNA restore defects in prostate most cancers. Eur. Urol. 76, 469–478 (2019).
Kumar, A. et al. Substantial interindividual and restricted intraindividual genomic range amongst tumors from males with metastatic prostate most cancers. Nat. Med. 22, 369–378 (2016).
Alshalalfa, M. et al. Clinicogenomic characterization of prostate most cancers liver metastases. Prostate Most cancers Prostatic Dis. 25, 366–369 (2022).
Samarzija, I. Website-specific and customary prostate most cancers metastasis genes as urged by meta-analysis of gene expression information. Life 11, 636 (2021).
Faltermeier, C. M. et al. Practical display identifies kinases driving prostate most cancers visceral and bone metastasis. Proc. Natl Acad. Sci. USA 113, E172–E181 (2016).
Lee, J. W. et al. Hepatocytes direct the formation of a pro-metastatic area of interest within the liver. Nature 567, 249–252 (2019).
Eveno, C. et al. Proof of prometastatic area of interest induction by hepatic stellate cells. J. Surg. Res. 194, 496–504 (2015).
Kopp, W. [Morphometric distribution of HLA-DR-coded immunocytes in gingiva biopsy specimens with periodontal disease]. Dtsch. Zahnarztl. Z. 45, 93–97 (1990).
Kondo, T. et al. The influence of hepatic fibrosis on the incidence of liver metastasis from colorectal most cancers. Br. J. Most cancers 115, 34–39 (2016).
Ohlund, D., Elyada, E. & Tuveson, D. Fibroblast heterogeneity within the most cancers wound. J. Exp. Med. 211, 1503–1523 (2014).
Costa-Silva, B. et al. Pancreatic most cancers exosomes provoke pre-metastatic area of interest formation within the liver. Nat. Cell Biol. 17, 816–826 (2015).
Hintz, H. M. et al. Imaging fibroblast activation protein alpha improves analysis of metastatic prostate most cancers with positron emission tomography. Clin. Most cancers Res. 26, 4882–4891 (2020).
James, N. D. et al. Abiraterone for prostate most cancers not beforehand handled with hormone remedy. N. Engl. J. Med. 377, 338–351 (2017).
Colomba, E. et al. Liver assessments improve on abiraterone acetate in males with metastatic prostate most cancers: pure historical past, administration and end result. Eur. J. Most cancers 129, 117–122 (2020).
Yun, G. Y. et al. Atypical onset of bicalutamide-induced liver harm. World J. Gastroenterol. 22, 4062–4065 (2016).
Kim, O. H. et al. Fluid shear stress facilitates prostate most cancers metastasis by means of Piezo1-Src-YAP axis. Life Sci. 308, 120936 (2022).
Akoto, T. & Saini, S. Position of exosomes in prostate most cancers metastasis. Int. J. Mol. Sci. 22, 3528 (2021).
Vagner, T. et al. Giant extracellular vesicles carry many of the tumour DNA circulating in prostate most cancers affected person plasma. J. Extracell. Vesicles 7, 1505403 (2018).
Teng, Y. et al. MVP-mediated exosomal sorting of miR-193a promotes colon most cancers development. Nat. Commun. 8, 14448 (2017).
Li, S. et al. Tumour-derived exosomes in liver metastasis: a Pandora’s field. Cell Prolif. 56, e13452 (2023).
Hoshino, A. et al. Tumour exosome integrins decide organotropic metastasis. Nature 527, 329–335 (2015).
Li, C. et al. hnRNPA2B1-mediated extracellular vesicles sorting of miR-122-5p probably promotes lung most cancers development. Int. Mol. J. Sci. 22, 12866 (2021).
Zhang, C. et al. Most cancers-derived exosomal HSPC111 promotes colorectal most cancers liver metastasis by reprogramming lipid metabolism in cancer-associated fibroblasts. Cell Loss of life Dis. 13, 57 (2022).
Zeng, Z. et al. Most cancers-derived exosomal miR-25-3p promotes pre-metastatic area of interest formation by inducing vascular permeability and angiogenesis. Nat. Commun. 9, 5395 (2018).
Kim, J. et al. Three-dimensional human liver-chip emulating premetastatic area of interest formation by breast cancer-derived extracellular vesicles. ACS Nano 14, 14971–14988 (2020).
Novizio, N. et al. ANXA1 contained in EVs regulates macrophage polarization in tumor microenvironment and promotes pancreatic most cancers development and metastasis. Int. J. Mol. Sci. 22, 11018 (2021).
Baig, M. S. et al. Tumor-derived exosomes within the regulation of macrophage polarization. Inflamm. Res. 69, 435–451 (2020).
Chang, Y. T. et al. Pancreatic cancer-derived small extracellular vesical Ezrin regulates macrophage polarization and promotes metastasis. Am. J. Most cancers Res. 10, 12–37 (2020).
Yin, X. et al. MiR-26b-5p in small extracellular vesicles derived from dying tumor cells after irradiation enhances the metastasis selling microenvironment in esophageal squamous cell carcinoma. Most cancers Lett. 541, 215746 (2022).
Solar, H. et al. Hypoxia-inducible exosomes facilitate liver-tropic premetastatic area of interest in colorectal most cancers. Hepatology 74, 2633–2651 (2021).
Kmiec, Z. Cooperation of liver cells in well being and illness. Adv. Anat. Embryol. Cell Biol. 161, 1–151 (2001). III-XIII.
Matsumura, H. et al. Kupffer cells lower metastasis of colon most cancers cells to the liver within the early stage. Int. J. Oncol. 45, 2303–2310 (2014).
Condamine, T. & Gabrilovich, D. I. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and performance. Traits Immunol. 32, 19–25 (2011).
Zhao, W. et al. Hepatic stellate cells promote tumor development by enhancement of immunosuppressive cells in an orthotopic liver tumor mouse mannequin. Lab. Make investments. 94, 182–191 (2014).
Bu, P. et al. Aldolase B-mediated fructose metabolism drives metabolic reprogramming of colon most cancers liver metastasis. Cell Metab. 27, 1249–1262 e1244 (2018).
Spratt, D. E. et al. Utility of FDG-PET in scientific neuroendocrine prostate most cancers. Prostate 74, 1153–1159 (2014).
Yates, C. C., Shepard, C. R., Stolz, D. B. & Wells, A. Co-culturing human prostate carcinoma cells with hepatocytes results in elevated expression of E-cadherin. Br. J. Most cancers 96, 1246–1252 (2007).
Wells, A., Yates, C. & Shepard, C. R. E-cadherin as an indicator of mesenchymal to epithelial reverting transitions throughout the metastatic seeding of disseminated carcinomas. Clin. Exp. Metastasis 25, 621–628 (2008).
Ma, B. & Wells, A. The mitogen-activated protein (MAP) kinases p38 and extracellular signal-regulated kinase (ERK) are concerned in hepatocyte-mediated phenotypic switching in prostate most cancers cells. J. Biol. Chem. 289, 11153–11161 (2014).
Ma, B. et al. Liver protects metastatic prostate most cancers from induced loss of life by activating E-cadherin signaling. Hepatology 64, 1725–1742 (2016).
Ma, B., Khazali, A., Shao, H., Jiang, Y. & Wells, A. Expression of E-cadherin and particular CXCR3 isoforms influence one another in prostate most cancers. Cell Commun. Sign. 17, 164 (2019).
Lin, H. Y. et al. Matriptase-2/NR4A3 axis switches TGF-β motion towards suppression of prostate most cancers cell invasion, tumor development, and metastasis. Oncogene 41, 2833–2845 (2022).
Wu, Q., Dhir, R. & Wells, A. Altered CXCR3 isoform expression regulates prostate most cancers cell migration and invasion. Mol. Most cancers 11, 3 (2012).
Zhu, W. B., Zhao, Z. F. & Zhou, X. AMD3100 inhibits epithelial-mesenchymal transition, cell invasion, and metastasis within the liver and the lung by means of blocking the SDF-1ɑ/CXCR4 signaling pathway in prostate most cancers. J. Cell Physiol. 234, 11746–11759 (2019).
Steele, R., Mott, J. L. & Ray, R. B. MBP-1 upregulates miR-29b that represses Mcl-1, collagens, and matrix-metalloproteinase-2 in prostate most cancers cells. Genes. Most cancers 1, 381–387 (2010).
Ru, P. et al. miRNA-29b suppresses prostate most cancers metastasis by regulating epithelial-mesenchymal transition signaling. Mol. Most cancers Ther. 11, 1166–1173 (2012).
Liu, C. et al. The microRNA miR-34a inhibits prostate most cancers stem cells and metastasis by straight repressing CD44. Nat. Med. 17, 211–215 (2011).
Tauriello, D. V. F. et al. TGFβ drives immune evasion in genetically reconstituted colon most cancers metastasis. Nature 554, 538–543 (2018).
Gao, Y. et al. Tumor immunoevasion by the conversion of effector NK cells into sort 1 innate lymphoid cells. Nat. Immunol. 18, 1004–1015 (2017).
Correia, A. L. et al. Hepatic stellate cells suppress NK cell-sustained breast most cancers dormancy. Nature 594, 566–571 (2021).
Ducimetiere, L. et al. Typical NK cells and tissue-resident ILC1s be part of forces to regulate liver metastasis. Proc. Natl Acad. Sci. USA 118, e2026271118 (2021).
Donkor, M. Okay. et al. T cell surveillance of oncogene-induced prostate most cancers is impeded by T cell-derived TGF-β1 cytokine. Immunity 35, 123–134 (2011).
Yu, J. et al. Liver metastasis restrains immunotherapy efficacy by way of macrophage-mediated T cell elimination. Nat. Med. 27, 152–164 (2021).
Ham, B. et al. TNF receptor-2 facilitates an immunosuppressive microenvironment within the liver to advertise the colonization and development of hepatic metastases. Most cancers Res. 75, 5235–5247 (2015).
Yang, S. et al. Built-in multi-omics panorama of liver metastases. Gastroenterology 164, 407–423 e417 (2023).
Wu, J. B. et al. MAOA-dependent activation of Shh-IL6-RANKL signaling community promotes prostate most cancers metastasis by partaking tumor-stromal cell interactions. Most cancers Cell 31, 368–382 (2017).
Wegiel, B. et al. A number of mobile mechanisms associated to cyclin A1 in prostate most cancers invasion and metastasis. J. Natl Most cancers Inst. 100, 1022–1036 (2008).
Klezovitch, O. et al. Hepsin promotes prostate most cancers development and metastasis. Most cancers Cell 6, 185–195 (2004).
Hsin, F., Hsu, Y. C., Tsai, Y. F., Lin, S. W. & Liu, H. M. The transmembrane serine protease hepsin suppresses sort I interferon induction by cleaving STING. Sci. Sign 14, eabb4752 (2021).
Su, W. et al. The polycomb repressor complicated 1 drives double-negative prostate most cancers metastasis by coordinating stemness and immune suppression. Most cancers Cell 36, 139–155 e110 (2019).
Sailer, V. et al. Experimental in vitro, ex vivo and in vivo fashions in prostate most cancers analysis. Nat. Rev. Urol. 20, 158–178 (2023).
Gao, D. et al. Organoid cultures derived from sufferers with superior prostate most cancers. Cell 159, 176–187 (2014).
Pauli, C. et al. Personalised in vitro and in vivo most cancers fashions to information precision drugs. Most cancers Discov. 7, 462–477 (2017).
Hsiao, A. Y. et al. Microfluidic system for formation of PC-3 prostate most cancers co-culture spheroids. Biomaterials 30, 3020–3027 (2009).
Bock, N. et al. In vitro engineering of a bone metastases mannequin permits for examine of the consequences of antiandrogen therapies in superior prostate most cancers. Sci. Adv. 7, eabg2564 (2021).
Lau, W. M. et al. Identification of potential elements selling osteotropism in breast most cancers: a possible function for CITED2. Int. J. Most cancers 126, 876–884 (2010).
Qi, X. et al. An oncogenic hepatocyte-induced orthotopic mouse mannequin of hepatocellular most cancers arising within the setting of hepatic irritation and fibrosis. J. Vis. Exp. https://doi.org/10.3791/59368 (2019).
Liu, Okay. et al. A novel mouse mannequin for liver metastasis of prostate most cancers reveals dynamic tumour-immune cell communication. Cell Prolif. 54, e13056 (2021).
Acevedo, V. D. et al. Inducible FGFR-1 activation results in irreversible prostate adenocarcinoma and an epithelial-to-mesenchymal transition. Most cancers Cell 12, 559–571 (2007).
Cho, H. et al. RapidCaP, a novel GEM mannequin for metastatic prostate most cancers evaluation and remedy, reveals myc as a driver of Pten-mutant metastasis. Most cancers Discov. 4, 318–333 (2014).
Taranda, J. et al. Mixed whole-organ imaging at single-cell decision and immunohistochemical evaluation of prostate most cancers and its liver and mind metastases. Cell Rep. 37, 110027 (2021).
Zaporowska-Stachowiak, I. et al. Managing metastatic bone ache: new views, totally different options. Biomed. Pharmacother. 93, 1277–1284 (2017).
Alderton, G. Okay. Metastasis: instructions to metastatic websites. Nat. Rev. Most cancers 15, 696–697 (2015).
Xu, N. et al. Threat elements of creating visceral metastases at analysis in prostate most cancers sufferers. Transl. Most cancers Res. 8, 928–938 (2019).
Steffens, J., Friedmann, W. & Lobeck, H. Immunohistochemical analysis of the metastasizing prostatic carcinoma. Eur. Urol. 11, 91–94 (1985).
Whitney, C. A. et al. In males with castration-resistant prostate most cancers, visceral metastases predict shorter general survival: what predicts visceral metastases? Outcomes from the SEARCH database. Eur. Urol. Focus. 3, 480–486 (2017).
Minami, Y. & Kudo, M. Hepatic malignancies: correlation between sonographic findings and pathological options. World J. Radiol. 2, 249–256 (2010).
Sahani, D. V., Bajwa, M. A., Andrabi, Y., Bajpai, S. & Cusack, J. C. Present standing of imaging and rising strategies to guage liver metastases from colorectal carcinoma. Ann. Surg. 259, 861–872 (2014).
Tanaka, T. et al. Present imaging strategies for and imaging spectrum of prostate most cancers recurrence and metastasis: a pictorial evaluation. Radiographics 40, 709–726 (2020).
Namasivayam, S., Martin, D. R. & Saini, S. Imaging of liver metastases: MRI. Most cancers Imaging 7, 2–9 (2007).
Niekel, M. C., Bipat, S. & Stoker, J. Diagnostic imaging of colorectal liver metastases with CT, MR imaging, FDG PET, and/or FDG PET/CT: a meta-analysis of potential research together with sufferers who haven’t beforehand undergone therapy. Radiology 257, 674–684 (2010).
Floriani, I. et al. Efficiency of imaging modalities in analysis of liver metastases from colorectal most cancers: a scientific evaluation and meta-analysis. J. Magn. Reson. Imaging 31, 19–31 (2010).
Brimo, F. & Epstein, J. I. Immunohistochemical pitfalls in prostate pathology. Hum. Pathol. 43, 313–324 (2012).
de Galiza Barbosa, F. et al. Nonprostatic ailments on PSMA PET imaging: a spectrum of benign and malignant findings. Most cancers Imaging 20, 23 (2020).
Jadvar, H. Molecular imaging of prostate most cancers: PET radiotracers.AJR Am. J. Roentgenol. 199, 278–291 (2012).
Jadvar, H. Positron emission tomography in prostate most cancers: abstract of systematic critiques and meta-analysis. Tomography 1, 18–22 (2015).
Vaz, C. V. et al. Androgens improve the glycolytic metabolism and lactate export in prostate most cancers cells by modulating the expression of GLUT1, GLUT3, PFK, LDH and MCT4 genes. J. Most cancers Res. Clin. Oncol. 142, 5–16 (2016).
Jadvar, H. Molecular imaging of prostate most cancers with 18F-fluorodeoxyglucose PET. Nat. Rev. Urol. 6, 317–323 (2009).
Umbehr, M. H., Muntener, M., Hany, T., Sulser, T. & Bachmann, L. M. The function of 11C-choline and 18F-fluorocholine positron emission tomography (PET) and PET/CT in prostate most cancers: a scientific evaluation and meta-analysis. Eur. Urol. 64, 106–117 (2013).
Garcia, J. R. et al. [Low diagnostic yield of the 11C-choline PET/CT in the detection of liver metastasis from prostate cancer]. Rev. Esp. Med. Nucl. Imagen Mol. 33, 56–57 (2014).
Ghedini, P. et al. Liver metastases from prostate most cancers at 11C-Choline PET/CT: a multicenter, retrospective evaluation. Eur. J. Nucl. Med. Mol. Imaging 45, 751–758 (2018).
Bianchi, D., Rizzo, A., Bonacina, M., Zaniboni, A. & Savelli, G. Penile metastasis from prostate most cancers detected by 18F-fluorocholine PET/CT. Clin. Nucl. Med. 46, e38–e39 (2021).
Dejust, S., Messaoud, L., Jallerat, P., Marical, V. & Morland, D. Hepatic metastases from prostatic adenocarcinoma with out elevated 18F-choline exercise. Clin. Nucl. Med. 43, 780–781 (2018).
Onner, H., Ozer, H., Celik, A. V., Yilmaz, F. & Kara Gedik, G. Remoted liver metastasis detected by 68Ga-PSMA PET/CT in newly recognized prostate most cancers. Clin. Nucl. Med. 48, 259–260 (2023).
De Man, Okay. et al. 18F-PSMA-11 versus 68Ga-PSMA-11 positron emission tomography/computed tomography for staging and biochemical recurrence of prostate most cancers: a potential double-blind randomised cross-over trial. Eur. Urol. 82, 501–509 (2022).
Seniaray, N., Verma, R., Belho, E., Malik, D. & Mahajan, H. Diffuse pulmonary metastases from prostate most cancers on 68Ga PSMA PET/CT. Clin. Nucl. Med. 44, 898–900 (2019).
Soydal, C., Ozkan, E., Yerlikaya, H., Utkan, G. & Kucuk, O. N. Widespread metastatic prostate carcinoma proven by 68Ga-PSMA PET/CT. Clin. Nucl. Med. 41, e294–e295 (2016).
Chan, M., Hsiao, E. & Turner, J. Cerebellar metastases from prostate most cancers on 68Ga-PSMA PET/CT. Clin. Nucl. Med. 42, 193–194 (2017).
Seifert, R. et al. Second model of the prostate most cancers molecular imaging standardized analysis framework together with response analysis for scientific trials (PROMISE V2). Eur. Urol. 83, 405–412 (2023).
Watt, F. et al. A tissue-specific enhancer of the prostate-specific membrane antigen gene, FOLH1. Genomics 73, 243–254 (2001).
Noss, Okay. R., Wolfe, S. A. & Grimes, S. R. Upregulation of prostate particular membrane antigen/folate hydrolase transcription by an enhancer. Gene 285, 247–256 (2002).
Beltran, H. et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate most cancers. Nat. Med. 22, 298–305 (2016).
Bakht, M. Okay. et al. Neuroendocrine differentiation of prostate most cancers results in PSMA suppression. Endocr. Relat. Most cancers 26, 131–146 (2018).
Thang, S. P. et al. Poor outcomes for sufferers with metastatic castration-resistant prostate most cancers with low prostate-specific membrane antigen (PSMA) expression deemed ineligible for 177Lu-labelled PSMA radioligand remedy. Eur. Urol. Oncol. 2, 670–676 (2019).
Evans, M. J. et al. Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen. Proc. Natl Acad. Sci. USA 108, 9578–9582 (2011).
Shetty, D., Patel, D., Le, Okay., Bui, C. & Mansberg, R. Pitfalls in gallium-68 PSMA PET/CT interpretation-A pictorial evaluation. Tomography 4, 182–193 (2018).
Ladron-de-Guevara, D., Canelo, A., Piottante, A. & Regonesi, C. False-positive 18F-prostate-specific membrane antigen-1007 PET/CT brought on by hepatic multifocal inflammatory foci. Clin. Nucl. Med. 46, e80–e83 (2021).
Kesch, C. et al. Excessive fibroblast-activation-protein expression in castration-resistant prostate most cancers helps using FAPI-molecular theranostics. Eur. J. Nucl. Med. Mol. Imaging 49, 385–389 (2021).
Kessel, Okay. et al. Prostate-specific membrane antigen and fibroblast activation protein distribution in prostate most cancers: preliminary information on immunohistochemistry and PET imaging. Ann. Nucl. Med. 36, 293–301 (2022).
Bluemel, C. et al. 68Ga-PSMA-PET/CT in sufferers with biochemical prostate most cancers recurrence and destructive 18F-Choline-PET/CT. Clin. Nucl. Med. 41, 515–521 (2016).
Bakht, M. Okay. et al. Differential expression of glucose transporters and hexokinases in prostate most cancers with a neuroendocrine gene signature: a mechanistic perspective for 18F-FDG imaging of PSMA-suppressed tumors. J. Nucl. Med. 61, 904–910 (2020).
Jadvar, H. The VISION ahead: recognition and implication of PSMA-/18F-FDG+ mCRPC. J. Nucl. Med. 63, 812–815 (2022).
Ignatiadis, M., Sledge, G. W. & Jeffrey, S. S. Liquid biopsy enters the clinic – implementation points and future challenges. Nat. Rev. Clin. Oncol. 18, 297–312 (2021).
Xu, L. et al. The novel affiliation of circulating tumor cells and circulating megakaryocytes with prostate most cancers prognosis. Clin. Most cancers Res. 23, 5112–5122 (2017).
Aceto, N. et al. Circulating tumor cell clusters are oligoclonal precursors of breast most cancers metastasis. Cell 158, 1110–1122 (2014).
Hille, C. & Pantel, Okay. Prostate most cancers: circulating tumour cells in prostate most cancers. Nat. Rev. Urol. 15, 265–266 (2018).
Chen, J. F. et al. Subclassification of prostate most cancers circulating tumor cells by nuclear measurement reveals very small nuclear circulating tumor cells in sufferers with visceral metastases. Most cancers 121, 3240–3251 (2015).
Snyder, M. W., Kircher, M., Hill, A. J., Daza, R. M. & Shendure, J. Cell-free DNA includes an in vivo nucleosome footprint that informs its tissues-of-origin. Cell 164, 57–68 (2016).
Mouliere, F. et al. Enhanced detection of circulating tumor DNA by fragment measurement evaluation. Sci. Transl. Med. 10, eaat4921 (2018).
Mehra, N. et al. Plasma cell-free DNA focus and outcomes from taxane remedy in metastatic castration-resistant prostate most cancers from two section III trials (FIRSTANA and PROSELICA). Eur. Urol. 74, 283–291 (2018).
Vandekerkhove, G. et al. Circulating tumor DNA abundance and potential utility in de novo metastatic prostate most cancers. Eur. Urol. 75, 667–675 (2019).
Wyatt, A. W. et al. Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate most cancers. J. Natl Most cancers Inst. 109, djx118 (2017).
Schweizer, M. T. et al. Concordance of DNA restore gene mutations in paired main prostate most cancers samples and metastatic tissue or cell-free DNA. JAMA Oncol. 7, 1–5 (2021).
Beltran, H. et al. Circulating tumor DNA profile acknowledges transformation to castration-resistant neuroendocrine prostate most cancers. J. Clin. Make investments. 130, 1653–1668 (2020).
Del Re, M. et al. The detection of androgen receptor splice variant 7 in plasma-derived exosomal RNA strongly predicts resistance to hormonal remedy in metastatic prostate most cancers sufferers. Eur. Urol. 71, 680–687 (2017).
Zhang, Y. et al. Lack of exosomal miR-146a-5p from cancer-associated fibroblasts after androgen deprivation remedy contributes to prostate most cancers metastasis. J. Exp. Clin. Most cancers Res. 39, 282 (2020).
Bhagirath, D. et al. microRNA-1246 is an exosomal biomarker for aggressive prostate most cancers. Most cancers Res. 78, 1833–1844 (2018).
Davis, I. D. et al. Enzalutamide with commonplace first-line remedy in metastatic prostate most cancers. N. Engl. J. Med. 381, 121–131 (2019).
Goodman, O. B. Jr et al. Exploratory evaluation of the visceral illness subgroup in a section III examine of abiraterone acetate in metastatic castration-resistant prostate most cancers. Prostate Most cancers Prostatic Dis. 17, 34–39 (2014).
Berthold, D. R. et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for superior prostate most cancers: up to date survival within the TAX 327 examine. J. Clin. Oncol. 26, 242–245 (2008).
de Wit, R. et al. Cabazitaxel versus abiraterone or enzalutamide in metastatic prostate most cancers. N. Engl. J. Med. 381, 2506–2518 (2019).
Alabi, B. R., Liu, S. & Stoyanova, T. Present and rising therapies for neuroendocrine prostate most cancers. Pharmacol. Ther. 238, 108255 (2022).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT04709276 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05413421 (2024).
Fizazi, Okay. et al. Abiraterone plus prednisone added to androgen deprivation remedy and docetaxel in de novo metastatic castration-sensitive prostate most cancers (PEACE-1): a multicentre, open-label, randomised, section 3 examine with a 2 x 2 factorial design. Lancet 399, 1695–1707 (2022).
Hussain, M. et al. Darolutamide plus androgen-deprivation remedy and docetaxel in metastatic hormone-sensitive prostate most cancers by illness quantity and danger subgroups within the section III ARASENS trial. J. Clin. Oncol. 41, 3595–3607 (2023).
Clarke, N. W. Abiraterone and olaparib for metastatic castration-resistant prostate most cancers. N. Engl. J. Med. Evid. 1, 9 (2022).
Agarwal, N. et al. Talazoparib plus enzalutamide in males with first-line metastatic castration-resistant prostate most cancers (TALAPRO-2): a randomised, placebo-controlled, section 3 trial. Lancet 402, 291–303 (2023).
de Bono, J. S. et al. Talazoparib monotherapy in metastatic castration-resistant prostate most cancers with DNA restore alterations (TALAPRO-1): an open-label, section 2 trial. Lancet Oncol. 22, 1250–1264 (2021).
Chi, Okay. N. et al. Niraparib plus abiraterone acetate with prednisone in sufferers with metastatic castration-resistant prostate most cancers and homologous recombination restore gene alterations: second interim evaluation of the randomized section III MAGNITUDE trial. Ann. Oncol. 34, 772–782 (2023).
Abida, W. et al. Rucaparib in males with metastatic castration-resistant prostate most cancers harboring a BRCA1 or BRCA2 gene alteration. J. Clin. Oncol. 38, 3763–3772 (2020).
Petrylak, D. P. et al. Pembrolizumab plus docetaxel for sufferers with metastatic castration-resistant prostate most cancers (mCRPC): randomized, double-blind, section 3 KEYNOTE-921 examine. J. Clin. Oncol. 41, 19–19 (2023).
Hennrich, U. & Eder, M. [177Lu]Lu-PSMA-617 (PluvictoTM): the primary FDA-approved radiotherapeutical for therapy of prostate most cancers. Prescribed drugs 15, 1292 (2022).
Sartor, O. et al. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate most cancers. N. Engl. J. Med. 385, 1091–1103 (2021).
Pouessel, D. et al. Liver metastases in prostate carcinoma: scientific traits and end result. BJU Int. 99, 807–811 (2007).
Ferdinandus, J. et al. Predictors of response to radioligand remedy of metastatic castrate-resistant prostate most cancers with 177Lu-PSMA-617. J. Nucl. Med. 58, 312–319 (2017).
Violet, J. et al. Dosimetry of 177Lu-PSMA-617 in metastatic castration-resistant prostate most cancers: correlations between pretherapeutic imaging and whole-body tumor dosimetry with therapy outcomes. J. Nucl. Med. 60, 517–523 (2019).
Park, I. Okay. et al. Impact of equine-assisted actions on cardiac autonomic operate in youngsters with cerebral palsy: a pilot randomized-controlled trial. J. Altern. Complement. Med. 27, 96–102 (2021).
Kyte, J. A. Most cancers vaccination with telomerase peptide GV1001. Professional. Opin. Investig. Medication 18, 687–694 (2009).
Kim, J. W. et al. Anti-metastatic impact of GV1001 on prostate most cancers cells; roles of GnRHR-mediated Gɑs-cAMP pathway and AR-YAP1 axis. Cell Biosci. 11, 191 (2021).
Agarwal, N. et al. Cabozantinib together with atezolizumab in sufferers with metastatic castration-resistant prostate most cancers: outcomes from an growth cohort of a multicentre, open-label, section 1b trial (COSMIC-021). Lancet Oncol. 23, 899–909 (2022).
Agarwal, N. et al. A section III, randomized, open-label examine (CONTACT-02) of cabozantinib plus atezolizumab versus second novel hormone remedy in sufferers with metastatic castration-resistant prostate most cancers. Future Oncol. 18, 1185–1198 (2022).
Pang, Y. et al. Improvement of FAPI tetramers to enhance tumor uptake and efficacy of FAPI radioligand remedy. J. Nucl. Med. 64, 1449–1455 (2023).
Feuerecker, B. et al. Exercise and antagonistic occasions of actinium-225-PSMA-617 in superior metastatic castration-resistant prostate most cancers after failure of lutetium-177-PSMA. Eur. Urol. 79, 343–350 (2021).
Ballal, S. et al. Lengthy-term survival outcomes of salvage [225Ac]Ac-PSMA-617 focused alpha remedy in sufferers with PSMA-expressing end-stage metastatic castration-resistant prostate most cancers: a real-world examine. Eur. J. Nucl. Med. Mol. Imaging 50, 3777–3789 (2023).
Sathekge, M. M., Bruchertseifer, F., Vorster, M., Morgenstern, A. & Lawal, I. O. World expertise with PSMA-based alpha remedy in prostate most cancers. Eur. J. Nucl. Med. Mol. Imaging 49, 30–46 (2021).
Zang, J. et al. First-in-human examine of 177Lu-EB-PSMA-617 in sufferers with metastatic castration-resistant prostate most cancers. Eur. J. Nucl. Med. Mol. Imaging 46, 148–158 (2019).
Wang, G. et al. A single-arm, low-dose, potential examine of 177Lu-EB-PSMA radioligand remedy in sufferers with metastatic castration-resistant prostate most cancers. J. Nucl. Med. 64, 611–617 (2023).
Ost, P. et al. Metastasis-directed remedy of regional and distant recurrences after healing therapy of prostate most cancers: a scientific evaluation of the literature. Eur. Urol. 67, 852–863 (2015).
Wang, S. C., McCarthy, L. P. & Mehdi, S. Remoted hepatic metastasis from prostate carcinoma. Urol. Case Rep. 10, 51–53 (2017).
Battaglia, A. et al. Metastasectomy for visceral and skeletal oligorecurrent prostate most cancers. World J. Urol. 37, 1543–1549 (2019).
Zager, J. S. et al. FOCUS section 3 trial outcomes: percutaneous hepatic perfusion (PHP) with melphalan for sufferers with ocular melanoma liver metastases (PHP-OCM-301/301A). J. Clin. Oncol. 40, 9510–9510 (2022).
Chalkidou, A. et al. Stereotactic ablative physique radiotherapy in sufferers with oligometastatic cancers: a potential, registry-based, single-arm, observational, analysis examine. Lancet Oncol. 22, 98–106 (2021).
Wang, H., Li, X., Peng, R., Wang, Y. & Wang, J. Stereotactic ablative radiotherapy for colorectal most cancers liver metastasis. Semin. Most cancers Biol. 71, 21–32 (2021).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT02239900 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT02710253 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT02843165 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT02888743 (2024).
Corn, P. G. et al. Cabazitaxel plus carboplatin for the therapy of males with metastatic castration-resistant prostate cancers: a randomised, open-label, section 1–2 trial. Lancet Oncol. 20, 1432–1443 (2019).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT04592237 (2024).
Blum, A., Wang, P. & Zenklusen, J. C. SnapShot: TCGA analyzed tumors. Cell 173, 530 (2018).
Cerami, E. et al. The cBio most cancers genomics portal: an open platform for exploring multidimensional most cancers genomics information. Most cancers Discov. 2, 401–404 (2012).
Robinson, D. et al. Integrative scientific genomics of superior prostate most cancers. Cell 161, 1215–1228 (2015).
Fizazi, Okay. et al. Well being-related high quality of life and ache outcomes with [177Lu]Lu-PSMA-617 plus commonplace of care versus commonplace of care in sufferers with metastatic castration-resistant prostate most cancers (VISION): a multicentre, open-label, randomised, section 3 trial. Lancet Oncol. 24, 597–610 (2023).
Chi, Okay. N. et al. Niraparib and abiraterone acetate for metastatic castration-resistant prostate most cancers. J. Clin. Oncol. 41, 3339–3351 (2023).
Kelly, W. Okay. et al. Randomized, double-blind, placebo-controlled section III trial evaluating docetaxel and prednisone with or with out bevacizumab in males with metastatic castration-resistant prostate most cancers: CALGB 90401. J. Clin. Oncol. 30, 1534–1540 (2012).
Chi, Okay. N. et al. Custirsen together with docetaxel and prednisone for sufferers with metastatic castration-resistant prostate most cancers (SYNERGY trial): a section 3, multicentre, open-label, randomised trial. Lancet Oncol. 18, 473–485 (2017).
Saad, F. et al. Orteronel plus prednisone in sufferers with chemotherapy-naive metastatic castration-resistant prostate most cancers (ELM-PC 4): a double-blind, multicentre, section 3, randomised, placebo-controlled trial. Lancet Oncol. 16, 338–348 (2015).
Oudard, S. et al. Cabazitaxel versus docetaxel as first-line remedy for sufferers with metastatic castration-resistant prostate most cancers: a randomized section III trial-FIRSTANA. J. Clin. Oncol. 35, 3189–3197 (2017).
Vogelzang, N. J. et al. Efficacy and security of autologous dendritic cell-based immunotherapy, docetaxel, and prednisone vs placebo in sufferers with metastatic castration-resistant prostate most cancers: the VIABLE section 3 randomized scientific trial. JAMA Oncol. 8, 546–552 (2022).
Saad, F. et al. Apalutamide plus abiraterone acetate and prednisone versus placebo plus abiraterone and prednisone in metastatic, castration-resistant prostate most cancers (ACIS): a randomised, placebo-controlled, double-blind, multinational, section 3 examine. Lancet Oncol. 22, 1541–1559 (2021).
Morris, M. J. et al. Randomized section III examine of enzalutamide in contrast with enzalutamide plus abiraterone for metastatic castration-resistant prostate most cancers (Alliance A031201 Trial). J. Clin. Oncol. 41, 3352–3362 (2023).
Kellokumpu-Lehtinen, P. L. et al. 2-Weekly versus 3-weekly docetaxel to deal with castration-resistant superior prostate most cancers: a randomised, section 3 trial. Lancet Oncol. 14, 117–124 (2013).
Beer, T. M. et al. Custirsen (OGX-011) mixed with cabazitaxel and prednisone versus cabazitaxel and prednisone alone in sufferers with metastatic castration-resistant prostate most cancers beforehand handled with docetaxel (AFFINITY): a randomised, open-label, worldwide, section 3 trial. Lancet Oncol. 18, 1532–1542 (2017).
Fizazi, Okay. et al. Part III, randomized, double-blind, multicenter trial evaluating orteronel (TAK-700) plus prednisone with placebo plus prednisone in sufferers with metastatic castration-resistant prostate most cancers that has progressed throughout or after docetaxel-based remedy: ELM-PC 5. J. Clin. Oncol. 33, 723–731 (2015).
Eisenberger, M. et al. Part III examine evaluating a diminished dose of cabazitaxel (20 mg/m2) and the at the moment authorized dose (25 mg/m2) in postdocetaxel sufferers with metastatic castration-resistant prostate cancer-PROSELICA. J. Clin. Oncol. 35, 3198–3206 (2017).
Antonarakis, E. S. et al. Pembrolizumab plus olaparib for sufferers with beforehand handled and biomarker-unselected metastatic castration-resistant prostate most cancers: the randomized, open-label, section III KEYLYNK-010 trial. J. Clin. Oncol. 41, 3839–3850 (2023).
Smith, M. et al. Part III examine of cabozantinib in beforehand handled metastatic castration-resistant prostate most cancers: COMET-1. J. Clin. Oncol. 34, 3005–3013 (2016).
Gravis, G. et al. Androgen-deprivation remedy alone or with docetaxel in non-castrate metastatic prostate most cancers (GETUG-AFU 15): a randomised, open-label, section 3 trial. Lancet Oncol. 14, 149–158 (2013).
Chi, Okay. N. et al. Apalutamide for metastatic, castration-sensitive prostate most cancers. N. Engl. J. Med. 381, 13–24 (2019).
Agarwal, N. et al. Orteronel for metastatic hormone-sensitive prostate most cancers: a multicenter, randomized, open-label section III trial (SWOG-1216). J. Clin. Oncol. 40, 3301–3309 (2022).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT03179410 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT03582475 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT03910660 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT03896503 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT04702737 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT04848337 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT04926181 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05582031 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05605522 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05652686 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05988918 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT05691465 (2024).
US Nationwide Library of Medication. ClinicalTrials.gov https://clinicaltrials.gov/examine/NCT06094842 (2024).