Skip to main content

OPINION article

Front. Oncol., 22 May 2018
Sec. Genitourinary Oncology
This article is part of the Research Topic Response and Resistance in Castration-Resistant Prostate Cancer View all 10 articles

Supraphysiological Testosterone Therapy as Treatment for Castration-Resistant Prostate Cancer

  • Department of Urology, University of Washington School of Medicine, Seattle, WA, United States

Blocking androgen signaling has been the focus of treatment for advanced and metastatic prostate cancer (PC) for the past 70 years (1). First-line androgen deprivation therapy (ADT), either through surgical or medical castration (luteinizing hormone-releasing hormone agonists and antiandrogens), holds promise for PC patients; however, the disease inevitably progresses to castration resistance (2). Second-generation ADT, abiraterone acetate (AA), and enzalutamide (ENZ), have been effective for a subset of patients with castration-resistant PC (CRPC) with relatively short median survival benefits (~3–5 months) (35). Concerted effort in the field, including evidence from our group, clearly demonstrates a sustained AR activity in the CRPC tumors including (1) amplification of AR, (2) AR mutations, (3) expression of AR splice variants that are constitutively active, (4) altered milieu of AR coactivators and corepressors, and (5) intracrine synthesis of androgens to support CRPC progression (6, 7).

The addiction of PC to the AR signaling paradoxically creates a therapeutic vulnerability that has recently attracted increasing attention. While ADT causes regression of PC, high level of androgen can also inhibit PC progression. The concept of cancer suppression using excessive hormone therapy was introduced by earlier work from Huggins in 1940: “malignant cells can regress from too little or too much hormone” (8). In relation to PC, AR regulates proliferation as well as differentiation of prostate epithelial and cancer cells but it has not been established what conditions support one over the other. Interestingly activation of AR with excessive hormone (i.e., supraphysiological levels of testosterone; SPT) was shown to inhibit growth of CRPC in vitro by negative effects on proliferation and increased expression of some of the AR-regulated genes that are expressed in differentiated luminal epithelium, e.g., prostate-specific antigen. Multiple preclinical studies demonstrated that SPT inhibits growth of PC cells that express AR (921), with evidence suggesting that higher levels of AR might lead to more pronounced SPT effects in certain phenotypes of CRPC [reviewed in Ref. (22)]. However, AR by itself is not necessarily sufficient for the SPT-induced growth inhibition; cellular context (23) and AR-regulated transcriptome in its entirety will need to be assessed to delineate the molecular effect of SPT (24). Mechanistically, SPT-induced cell growth inhibition involves (1) cell-cycle arrest, (2) disruption of AR-mediated DNA licensing, (3) DNA damage, (4) transcriptional repression of AR and its variants, (5) transcriptional reprogramming, (6) cellular quiescence or senescence, and (7) induction of apoptosis [reviewed in Ref. (22)]. However, these effects were demonstrated exclusively in cell line models, and whether they play a significant biological role in SPT-induced tumor inhibition in patients remains to be determined.

Clinical use of testosterone (T) supplementation in PC has been limited and provided controversial results. Two older National Prostatic Cancer Project trials that used T-supplementation to normal levels with a goal to enhance the effectiveness of chemotherapy reported disappointing results (25, 26). Additional two phase I trials, which did not achieve consistent supraphysiological T levels, showed minimally reduced disease progression (27, 28). In contrast, several other studies showed that T-supplementation to normal-supraphysiological range (303−2637 ng/dl), specifically in symptomatic hypogonadal PC patients, provided prolonged disease control (as measured by sustainably low-PSA level) (2931). In our opinion, the lack of favorable response in some of the clinical trials is, at least in part, due to the absence of a supraphysiological level of T as well as the unselected patient population.

With advanced understanding of the biology and AR involvement in CRPC progression, leveraging the active AR signaling to explore therapeutic opportunity has recently received renewed attention in clinical settings. Dr. Denmeade’s group at John Hopkins University pioneered a therapy termed “bipolar androgen therapy” (BAT) as a treatment for PC patients. With BAT treatment, PC patients receive intermittent T injections at doses shown to produce a spike in serum T to supraphysiological levels, followed by a decline to below normal at the end of a 28-day treatment cycle (32). This cycling strategy was developed based on the most common molecular hallmark of CRPC–overexpression of AR (33) and the potential growth inhibitory effect of SPT in AR-overexpressing PC. Rapidly cycling of T from SPT (~1,500 ng/dl) to below normal T levels (~150 ng/dl) was expected to blunt the adaptive changes in AR expression, thereby delaying the emergence of resistance. In these proof-of-principle BAT trials, one in CRPC showed radiographic response rates of ~50% in men (32), and one in hormone-sensitive PC showed favorable PSA responses (34). Promising results of these trials led to a new clinical trial, in which asymptomatic CRPC patients that progressed on AA or ENZ receive BAT, and after progression on BAT the patients are re-challenged with AA or ENZ. This trial aims to evaluate the efficacy of BAT in patients who progressed on secondary ADT and assess whether BAT re-sensitizes CRPC to secondary ADT. Recent data from this trial showed a PSA50 response in 9/30 ENZ-resistant patients on BAT, and, importantly, 15/21 patients who progressed on BAT showed a PSA50 response upon ENZ re-challenge (35). These results are encouraging. However, additional analyses and larger number of patients are needed to correlate tumor/radiographic vs. PSA responses in individual patients. One of the reasons is that PSA changes do not necessarily associate with tumor regression in advanced CRPC. PSA, an AR-regulated gene, is highly sensitive to AR activation/inhibition and can rise upon SPT and decline upon ADT. PSA response might not faithfully reflect radiographic responses in advanced CRPC which growth often does not rely solely on AR signaling [e.g., FGF signaling (36)].

Bipolar androgen therapy shows great clinical promise in a subset of patients. However, universal to all cancer treatment modalities, not all patients respond to this treatment and resistance to BAT develops. Therefore, there is an opportunity to improve this therapy. It is notable that a critical step in drug development, determining the optimal dosing schedule, was bypassed in the clinical development of BAT. Despite the clinical efficacy of BAT, there were by far no clinical data to support the hypotheses that cycling SPT (i.e., BAT) mitigates the development of resistance or that BAT represents the optimal mode for administering SPT. Notably, several preclinical studies have consistently demonstrated that SPT delivered on a continuous basis inhibits the growth of PC cells (13, 20, 21). While several small clinical trials of continuous T administration in men with CRPC have been carried out, they did not achieve SPT levels.

Cycling or not cycling—that is the question. While we currently do not have sufficient evidence whether BAT results in better clinical outcome than continuous SPT, it is possible that long-term continuous SPT and BAT could alter AR signaling differently. One would anticipate that continuous SPT might trigger more pronounced differentiation, potentially causing a change from a “low-T” oncogenic AR transcriptome to that of a more differentiating SPT transcriptome (24). Meanwhile, BAT might provide better efficacy if cell-cycle relicensing effects and DNA damage are the critical mechanism of action (37, 38). While BAT was associated with improved quality of life (34, 35), this effect diminished over the course of a cycle of BAT, presumably due to T levels falling below normal range. It is possible that quality of life metrics will be better with continuous SPT but there also might be increase in negative side effects. While T therapy has been reported to be generally safe, with a small subset of patients experiencing severe cardiovascular-related complications (2732, 3942), continuous SPT has not been tested and monitoring will be essential. Careful evaluation of effects of BAT vs. continuous SPT on tumor progression, as well as any potential health benefits or side effects will be required to make final decision.

Interestingly, cycling of ADT, intermittent ADT, has been evaluated in PC extensively since its introduction in mid-1980s. However, intermittent ADT was not found to be inferior to continuous ADT with respect to the overall survival but it was shown to improve patients’ quality of life, and therefore it is thought to be a viable option for patients who experience significant adverse effects of continuous ADT [for review see Ref. (43, 44)]. In addition, intermittent AA therapy has been recently shown to delay the development of resistance from 16.5 (continuous treatment) to 27 months (45). Whether the intermittent therapy diversifies the residual tumor clones or re-sensitize the residual clones to a therapy that formerly failed remains scientifically and clinically important.

In summary, we will need to seek answers to multiple important questions before unleashing the full potential of SPT therapy in CRPC: (1) which mode of SPT, BAT or continuous SPT, represents the optimal administration regimen for tumor growth inhibition; (2) what population of patients will benefit from SPT therapy; (3) is there a way to prolong the treatment response; and (4) what are the mechanisms of resistance, as these will be diverse in different tumor phenotypes. To address these questions, systematic preclinical trials will need to be performed, and pre-treatment and on-treatment clinical specimens will be essential to identify mechanisms of SPT action and biomarkers that predict SPT response.

Author Contributions

H-ML and EC jointly conceptualized and wrote the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Lisha Brown for critical reading of this manuscript.

Funding

This work was supported by NIH R21 CA194798, PNW Prostate Cancer SPORE P50 CA097186, P01 CA163227, and FHCRC/UW Cancer Consortium Cancer Center Support Grant P30 CA015704.

References

1. Huggins C, Scott WW, Hodges CV. Studies on prostatic cancer III. The effects of fever, of desoxycorticosterone and of estrogen on clinical patients with metastatic carcinoma of the prosate. J Urol (1941) 46:997–1006. doi:10.1016/S0022-5347(17)71004-X

CrossRef Full Text | Google Scholar

2. Pienta KJ, Bradley D. Mechanisms underlying the development of androgen-independent prostate cancer. Clin Cancer Res (2006) 12(6):1665–71. doi:10.1158/1078-0432.CCR-06-0067

CrossRef Full Text | Google Scholar

3. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med (2013) 368(2):138–48. doi:10.1056/NEJMoa1209096

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Reid AH, Attard G, Barrie E, de Bono JS. CYP17 inhibition as a hormonal strategy for prostate cancer. Nat Clin Pract Urol (2008) 5(11):610–20. doi:10.1038/ncpuro1237

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med (2012) 367(13):1187–97. doi:10.1056/NEJMoa1207506

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Buttigliero C, Tucci M, Bertaglia V, Vignani F, Bironzo P, Di Maio M, et al. Understanding and overcoming the mechanisms of primary and acquired resistance to abiraterone and enzalutamide in castration resistant prostate cancer. Cancer Treat Rev (2015) 41(10):884–92. doi:10.1016/j.ctrv.2015.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Silberstein JL, Taylor MN, Antonarakis ES. Novel insights into molecular indicators of response and resistance to modern androgen-axis therapies in prostate cancer. Curr Urol Rep (2016) 17(4):29. doi:10.1007/s11934-016-0584-4

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Huggins C. Two principles in endocrine therapy of cancers: hormone deprival and hormone interference. Cancer Res (1965) 25(7):1163–7.

Google Scholar

9. Attardi BJ, Burgenson J, Hild SA, Reel JR. Steroid hormonal regulation of growth, prostate specific antigen secretion, and transcription mediated by the mutated androgen receptor in CWR22Rv1 human prostate carcinoma cells. Mol Cell Endocrinol (2004) 222(1–2):121–32. doi:10.1016/j.mce.2004.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Zhau HY, Chang SM, Chen BQ, Wang Y, Zhang H, Kao C, et al. Androgen-repressed phenotype in human prostate cancer. Proc Natl Acad Sci U S A (1996) 93(26):15152–7. doi:10.1073/pnas.93.26.15152

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Kokontis J, Takakura K, Hay N, Liao S. Increased androgen receptor activity and altered c-myc expression in prostate cancer cells after long-term androgen deprivation. Cancer Res (1994) 54(6):1566–73.

PubMed Abstract | Google Scholar

12. Kokontis JM, Hay N, Liao S. Progression of LNCaP prostate tumor cells during androgen deprivation: hormone-independent growth, repression of proliferation by androgen, and role for p27Kip1 in androgen-induced cell cycle arrest. Mol Endocrinol (1998) 12(7):941–53. doi:10.1210/mend.12.7.0136

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kokontis JM, Lin HP, Jiang SS, Lin CY, Fukuchi J, Hiipakka RA, et al. Androgen suppresses the proliferation of androgen receptor-positive castration-resistant prostate cancer cells via inhibition of Cdk2, CyclinA, and Skp2. PLoS One (2014) 9(10):e109170. doi:10.1371/journal.pone.0109170

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hara T, Nakamura K, Araki H, Kusaka M, Yamaoka M. Enhanced androgen receptor signaling correlates with the androgen-refractory growth in a newly established MDA PCa 2b-hr human prostate cancer cell subline. Cancer Res (2003) 63(17):5622–8.

PubMed Abstract | Google Scholar

15. Sonnenschein C, Olea N, Pasanen ME, Soto AM. Negative controls of cell proliferation: human prostate cancer cells and androgens. Cancer Res (1989) 49(13):3474–81.

PubMed Abstract | Google Scholar

16. Geck P, Maffini MV, Szelei J, Sonnenschein C, Soto AM. Androgen-induced proliferative quiescence in prostate cancer cells: the role of AS3 as its mediator. Proc Natl Acad Sci U S A (2000) 97(18):10185–90. doi:10.1073/pnas.97.18.10185

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Geck P, Szelei J, Jimenez J, Lin TM, Sonnenschein C, Soto AM. Expression of novel genes linked to the androgen-induced, proliferative shutoff in prostate cancer cells. J Steroid Biochem Mol Biol (1997) 63(4–6):211–8. doi:10.1016/S0960-0760(97)00122-2

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Maffini MV, Geck P, Powell CE, Sonnenschein C, Soto AM. Mechanism of androgen action on cell proliferation: AS3 protein as a mediator of proliferative arrest in the rat prostate. Endocrinology (2002) 143(7):2708–14. doi:10.1210/endo.143.7.8899

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Chuu CP, Hiipakka RA, Fukuchi J, Kokontis JM, Liao S. Androgen causes growth suppression and reversion of androgen-independent prostate cancer xenografts to an androgen-stimulated phenotype in athymic mice. Cancer Res (2005) 65(6):2082–4. doi:10.1158/0008-5472.CAN-04-3992

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Chuu CP, Kokontis JM, Hiipakka RA, Fukuchi J, Lin HP, Lin CY, et al. Androgen suppresses proliferation of castration-resistant LNCaP 104-R2 prostate cancer cells through androgen receptor, Skp2, and c-Myc. Cancer Sci (2011) 102(11):2022–8. doi:10.1111/j.1349-7006.2011.02043.x

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chuu CP, Kokontis JM, Hiipakka RA, Fukuchi J, Lin HP, Lin CY, et al. Androgens as therapy for androgen receptor-positive castration-resistant prostate cancer. J Biomed Sci (2011) 18:63. doi:10.1186/1423-0127-18-63

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Mohammad OS, Nyquist MD, Schweizer MT, Balk SP, Corey E, Plymate S, et al. Supraphysiologic testosterone therapy in the treatment of prostate cancer: models, mechanisms and questions. Cancers (Basel) (2017) 9(12):E166. doi:10.3390/cancers9120166

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Litvinov IV, Antony L, Dalrymple SL, Becker R, Cheng L, Isaacs JT. PC3, but not DU145, human prostate cancer cells retain the coregulators required for tumor suppressor ability of androgen receptor. Prostate (2006) 66(12):1329–38. doi:10.1002/pros.20483

CrossRef Full Text | Google Scholar

24. Cai C, He HH, Gao S, Chen S, Yu Z, Gao Y, et al. Lysine-specific demethylase 1 has dual functions as a major regulator of androgen receptor transcriptional activity. Cell Rep (2014) 9(5):1618–27. doi:10.1016/j.celrep.2014.11.008

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Fowler JE Jr, Whitmore WF Jr. The response of metastatic adenocarcinoma of the prostate to exogenous testosterone. J Urol (1981) 126(3):372–5. doi:10.1016/S0022-5347(17)54531-0

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Fowler JE Jr, Whitmore WF Jr. Considerations for the use of testosterone with systemic chemotherapy in prostatic cancer. Cancer (1982) 49(7):1373–7. doi:10.1002/1097-0142(19820401)49:7<1373::AID-CNCR2820490712>3.0.CO;2-G

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Morris MJ, Huang D, Kelly WK, Slovin SF, Stephenson RD, Eicher C, et al. Phase 1 trial of high-dose exogenous testosterone in patients with castration-resistant metastatic prostate cancer. Eur Urol (2009) 56(2):237–44. doi:10.1016/j.eururo.2009.03.073

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Szmulewitz R, Mohile S, Posadas E, Kunnavakkam R, Karrison T, Manchen E, et al. A randomized phase 1 study of testosterone replacement for patients with low-risk castration-resistant prostate cancer. Eur Urol (2009) 56(1):97–103. doi:10.1016/j.eururo.2009.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Leibowitz RL, Dorff TB, Tucker S, Symanowski J, Vogelzang NJ. Testosterone replacement in prostate cancer survivors with hypogonadal symptoms. BJU Int (2010) 105(10):1397–401. doi:10.1111/j.1464-410X.2009.08980.x

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Mathew P. Prolonged control of progressive castration-resistant metastatic prostate cancer with testosterone replacement therapy: the case for a prospective trial. Ann Oncol (2008) 19(2):395–6. doi:10.1093/annonc/mdm568

CrossRef Full Text | Google Scholar

31. Sarosdy MF. Testosterone replacement for hypogonadism after treatment of early prostate cancer with brachytherapy. Cancer (2007) 109(3):536–41. doi:10.1002/cncr.22438

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Schweizer MT, Antonarakis ES, Wang H, Ajiboye AS, Spitz A, Cao H, et al. Effect of bipolar androgen therapy for asymptomatic men with castration-resistant prostate cancer: results from a pilot clinical study. Sci Transl Med (2015) 7(269):269ra262. doi:10.1126/scitranslmed.3010563

CrossRef Full Text | Google Scholar

33. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell (2015) 161(5):1215–28. doi:10.1016/j.cell.2015.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Schweizer MT, Wang H, Luber B, Nadal R, Spitz A, Rosen DM, et al. Bipolar androgen therapy for men with androgen ablation naive prostate cancer: results from the phase II BATMAN study. Prostate (2016) 76(13):1218–26. doi:10.1002/pros.23209

CrossRef Full Text | Google Scholar

35. Teply BA, Wang H, Luber B, Sullivan R, Rifkind I, Bruns A, et al. Bipolar androgen therapy in men with metastatic castration-resistant prostate cancer after progression on enzalutamide: an open-label, phase 2, multicohort study. Lancet Oncol (2017) 19(1):76–86. doi:10.1016/S1470-2045(17)30906-3

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Bluemn EG, Coleman IM, Lucas JM, Coleman RT, Hernandez-Lopez S, Tharakan R, et al. Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling. Cancer Cell (2017) 32(4):474–89.e476. doi:10.1016/j.ccell.2017.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Hedayati M, Haffner MC, Coulter JB, Raval RR, Zhang Y, Zhou H, et al. Androgen deprivation followed by acute androgen stimulation selectively sensitizes AR-positive prostate cancer cells to ionizing radiation. Clin Cancer Res (2016) 22(13):3310–9. doi:10.1158/1078-0432.CCR-15-1147

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Isaacs JT, D’Antonio JM, Chen S, Antony L, Dalrymple SP, Ndikuyeze GH, et al. Adaptive auto-regulation of androgen receptor provides a paradigm shifting rationale for bipolar androgen therapy (BAT) for castrate resistant human prostate cancer. Prostate (2012) 72(14):1491–505. doi:10.1002/pros.22504

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Leibowitz RL, Tucker SJ. Treatment of localized prostate cancer with intermittent triple androgen blockade: preliminary results in 110 consecutive patients. Oncologist (2001) 6(2):177–82. doi:10.1634/theoncologist.6-2-177

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Kaufman JM, Graydon RJ. Androgen replacement after curative radical prostatectomy for prostate cancer in hypogonadal men. J Urol (2004) 172(3):920–2. doi:10.1097/01.ju.0000136269.10161.32

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Agarwal PK, Oefelein MG. Testosterone replacement therapy after primary treatment for prostate cancer. J Urol (2005) 173(2):533–6. doi:10.1097/01.ju.0000143942.55896.64

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Khera M. Androgen replacement therapy after prostate cancer treatment. Curr Urol Rep (2010) 11(6):393–9. doi:10.1007/s11934-010-0143-3

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Magnan S, Zarychanski R, Pilote L, Bernier L, Shemilt M, Vigneault E, et al. Intermittent vs continuous androgen deprivation therapy for prostate cancer: a systematic review and meta-analysis. JAMA Oncol (2015) 1(9):1261–9. doi:10.1001/jamaoncol.2015.2895

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Park JC, Eisenberger MA. Intermittent androgen deprivation in prostate cancer: are we ready to quit? J Clin Oncol (2016) 34(3):211–4. doi:10.1200/JCO.2015.64.1019

CrossRef Full Text | Google Scholar

45. Zhang J, Cunningham JJ, Brown JS, Gatenby RA. Integrating evolutionary dynamics into treatment of metastatic castrate-resistant prostate cancer. Nat Commun (2017) 8(1):1816. doi:10.1038/s41467-017-01968-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: supraphysiological testosterone, bipolar androgen therapy, castration-resistant prostate cancer, enzalutamide, abiraterone

Citation: Lam H-M and Corey E (2018) Supraphysiological Testosterone Therapy as Treatment for Castration-Resistant Prostate Cancer. Front. Oncol. 8:167. doi: 10.3389/fonc.2018.00167

Received: 10 January 2018; Accepted: 01 May 2018;
Published: 22 May 2018

Edited by:

Fabio Grizzi, Humanitas Research Hospital, Italy

Reviewed by:

Momoe Itsumi, Tokyo Medical and Dental University, Japan
Benyi Li, University of Kansas Medical Center, United States

Copyright: © 2018 Lam and Corey. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Eva Corey, ecorey@uw.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.