Abstract
Purpose of Review
The development of HIV protease inhibitors (PIs) more than two decades ago heralded a new era in HIV care, changing the infection from universally-fatal to chronic but controllable. With the widespread use of PIs, there was a reduction in the incidence and mortality of HIV-associated malignancies. Studies later found these drugs to have promising direct antitumor effects.
Recent findings
PIs have a wide range of effects on several cellular pathways that are important for tumorigenesis and independent of inhibition of the HIV protease, including reducing angiogenesis and cell invasion, inhibition of the Akt pathway, induction of autophagy, and promotion of apoptosis. Among PIs, Nelfinavir appears to have the most potent and broad antineoplastic activities, and also affects replication of the oncogenic herpesviruses Kaposi sarcoma-associated herpesvirus and Epstein-Barr virus. Nelfinavir is being studied for the prevention and treatment of a wide range of malignancies in persons with and without HIV infection.
Summary
Nelfinavir and other PIs are safe, oral drugs that have promising antitumor properties, and may prove to play an important role in the prevention and treatment of several cancers. Additional insights into PIs’ mechanisms of action may lead to the development of novel cancer chemotherapy agents.
Keywords: HIV protease inhibitor, nelfinavir, cancer, Kaposi sarcoma, treatment
Introduction
Protease inhibitors (PIs) were specifically designed to inhibit the HIV aspartyl protease, but serendipitously were found to have effects that confer broad antineoplastic and antiviral activity. This led to the idea that PIs could be efficiently repositioned for cancer chemotherapy [1,2], especially in light of their oral bioavailability, wide clinical use, and minimal toxicity. In particular, nelfinavir, now rarely used for antiretroviral therapy (ART), has emerged as the most potent candidate PI for many cancers in patients with or without HIV infection. This review provides an update on the use of PIs for cancer, with an emphasis on nelfinavir, including information on the antitumor mechanisms that have been identified, completed and ongoing clinical trials, and priorities for future research.
Description of HIV protease inhibitors
Protease inhibitors were developed to block the maturation of the HIV virion, by inhibiting the viral aspartyl protease’s cleavage of precursor polyproteins into their functional forms. Using the HIV protease crystal structure, specific small molecule inhibitors were rationally designed, leading to FDA approval of saquinavir for ART in 1995. Since that time, 9 additional HIV PIs have been approved: ritonavir, indinavir, nelfinavir, amprenavir, lopinavir, atazanavir, fosamprenavir, tipranavir, and darunavir. The use of PIs, in combination with available reverse transcriptase inhibitors, rapidly became the standard of care, and ushered in the era of “highly active” ART. Immediately afterward, mortality due to HIV infection began to decline, the incidence of opportunistic infections plummeted, and the number of new cases of AIDS-defining cancers (especially Kaposi sarcoma, KS) was markedly reduced.
Nelfinavir
The PI nelfinavir mesylate was approved in 1997, initially at a dose of 750 mg thrice daily, and later at 1250 mg BID. Serious adverse effects of nelfinavir are uncommon, though up to 30% of patients experience dose-dependent diarrhea [3]. Nelfinavir has subsequently been shown to be less effective against HIV than ritonavir-boosted PIs, and is no longer widely used for ART [4].
Evidence for anti-cancer activity of PIs in HIV-infected patients
Co-incident with the widespread use of PIs came a series of case reports demonstrating the regression of KS with PI-based ART alone [5-13]. However, as potent new non-nucleoside reverse transcriptase inhibitors (NNRTIs) were introduced, case series suggested that regression of KS was dependent more on the potency of the regimens to suppress HIV replication and reconstitute T cell immunity than on the individual drugs used [14-16]. Similarly, no difference has been seen in the reduction of risk for incident KS among HIV patients between those receiving regimens containing NNRTIs vs. PIs [17,18], including one study with a small number of KS cases which specifically looked at nelfinavir [19]. Though it is clear that all effective ART substantially reduces the risk of AIDS-defining cancers, to date no adequately powered randomized trial has evaluated the relative benefits of nelfinavir or other PIs for the treatment or prevention of KS or other malignancies in HIV-infected individuals.
In vitro and preclinical in vivo studies relevant for oncology
PIs have a broad range of effects on human cells that cause toxicity [4], but also confer potent anticancer properties (Table 1). Despite their limited homology with the HIV protease, cellular proteases appear to be the primary targets of PIs that account for their antitumor activity, particularly the proteasome and matrix metalloproteases (MMPs). However, numerous additional activities of PIs have been appreciated. A landmark paper by Gills et al [22] found that multiple PIs had activity against all 60 cell lines in the NCI60 panel, with nelfinavir being most potent among the drugs tested, and described multiple potential mechanisms of action against cancer, including endoplasmic reticulum (ER) stress, autophagy, apoptosis, and inhibition of Akt signaling. A comprehensive computational analysis of structural protein interactions found 92 predicted cellular targets of nelfinavir, of which the 7 with the strongest binding affinities were aspartyl proteases [59]. The list of remaining targets was dominated by protein kinases such growth factor receptors that are upstream regulators of Akt, NFαB, and other signaling molecules that are known to be down-regulated by nelfinavir [40]. PIs also have effects that may be disadvantageous for cancer treatment, including increased expression of multidrug transporter ABCB1 (P-glycoprotein) in KS cells, which confers in vitro resistance to doxorubicin and paclitaxel [60]. Considerable variation in the cellular effects of individual PIs has been observed, which may be both concentration- and cell type-dependent, making it challenging to predict tumor-specific activities.
Table 1. Antitumor mechanisms of HIV protease inhibitors.
Host cell effect | Protease inhibitor(s) | Cell types | References |
---|---|---|---|
Inhibition of Akt | nelfinavir, ritonavir, lopinavir, saquinavir, amprenavir |
multiple myeloma, pituitary adenoma, non-small cell lung cancer, head and neck squamous cell carcinoma, urinary bladder carcinoma, prostate cancer, glioma, breast cancer, diffuse large B cell lymphoma |
[20-31] |
ER stress | nelfinavir, ritonavir, atazanavir |
cervical cancer, renal cancer, glioma, breast cancer, non small cell lung cancer, head and neck squamous cell carcinoma, ovarian cancer, liposarcoma, multiple myeloma |
[22,29,31-39] |
Autophagy | nelfinavir | non small cell lung cancer, breast cancer, prostate cancer, chronic lymphocytic leukemia |
[22,40-43] |
Apoptosis | nelfinavir, saquinavir, ritonavir, lopinavir |
KS cell lines, breast cancer, non small cell lung cancer, hepatocellular carcinoma, cervical cancer renal carcinoma, Adult T cell leukemia, T cell lymphoma, EBV- positive B cell lymphoblastoid, prostate, gliobastoma multiforme, Jurkat leukemia, multiple myeloma, ovarian cancer, neuroblastoma, melanoma, Burkitt lymphoma, fibrosarcoma, myeloid leukemia |
[21,22,24,26,32 34,36,44-54] |
Matrix metalloprotease inhibition |
indinavir, ritonavir, saquinavir, amprenavir |
Cervical carcinoma/CIN; HCC |
[55-57] |
Inhibition of NFkB |
ritonavir, saquinavir | KS cell lines, Adult T cell leukemia, EBV- positive B cell lymphoblastoid, neuroblastoma |
[44,46,49,52] |
Proteasome inhibition |
saquinavir, nelfinavir, lopinavir, ritonavir |
Prostate, multiple myeloma, melanoma, cervical cancer, breast cancer, head and neck squamous cell carcinoma, murine T lymphocytes, T lymphoma |
[24,25,29,31,47, 48,50,54,58] |
Angiogenesis and cell invasion
Initial anticancer studies of PIs found that indinavir and saquinavir were potent inhibitors of angiogenesis and tumor cell invasion in murine KS models by blocking activation of MMP2 [57], and subsequently that ritonavir and saquinavir inhibit MMP2 and MMP9 and block invasion of cervical intraepithelial neoplasia cells in an in vitro system [55]. Amprenavir was also shown to inhibit MMP2 in hepatocarcinoma cells and impair cell invasion and tumor xenograft growth in nude mice [56]. Additionally, PIs can block angiogenesis through down-regulation of signaling pathways, such as phoshphatidylinositol 3-kinase (PI3K)/Akt, which modulates the expression of vascular endothelial growth factor (VEGF) and numerous other factors involved in neovascularization [27,44,56,61].
Inhibition of Akt
PI3K/Akt signaling and downstream mediators, such as mammalian target of rapamycin (mTOR) and VEGF, contribute to oncogenesis through effects on multiple cellular processes, including proliferation, motility, angiogenesis, transformation, apoptosis/survival, and DNA repair (reviewed in [62,63]). Upregulation of PI3K/Akt signaling protects against apoptotic cell death, and thereby confers resistance to radiation and chemotherapy in a number of cancers [63]. PIs inhibit phosphorylation of Akt in multiple tumor cell lines [20-22,25]. Nelfinavir appears to be the most potent inhibitor of Akt among the PI class [22], although this varies by cell type [25]. In rapamycin-resistant diffuse large B cell lymphoma lines in which Akt activation was upregulated, the combination of rapamycin with nelfinavir or the Akt inhibitor MK-2206 resulted in synergistic cytotoxicity [30]. A promising observational study of HIV-infected patients showed that those taking nelfinavir-based ART showed lower levels of Akt phosphorylation in leukocytes compared with patients on ART without a PI or not receiving antivirals; furthermore, nelfinavir was not associated with increased radiation toxicity [64]. However, the level of Akt inhibition does not always correlate with antitumor activity, and in some model systems nelfinavir paradoxically activates Akt [29,48]. The precise mechanism by which PIs prevent Akt phosphorylation by PI3K is unknown, but may result from inhibition of upstream growth factors, induction of ER stress, or other effects [29,31].
Endoplasmic reticulum stress
When accumulation misfolded proteins or other stresses overwhelm the ER equilibrium, the unfolded protein response (UPR) is triggered, which results in the attenuation of protein translation and cell cycle arrest (reviewed in [65,66]). Nelfinavir and other PIs cause ER stress [22,29,31,35-39]. In liposarcoma and castration-resistant prostate cancer cell lines (which has a lipogenic phenotype), nelfinavir induces overwhelming ER stress by inhibition of site-2 protease, resulting in impaired processing and accumulation of sterol regulatory element binding protein-1 (SREBP-1) and activating transcription factor 6 [38,43,67]. This fits well with what is known about a known toxicity of PIs, liposdystrophy, which appears to result from increased levels of SREBP-1 [1].
Proper protein folding is facilitated by chaperones, and degradation of misfolded proteins is performed primarily by the proteasome. As such, interference with either of these functions by PIs may contribute to ER stress [24,29,35,39,53]. Inhibition of 20S proteasome activity by ritonavir [44,54,58] and saquinavir [50] has been reported. Nelfinavir also has cell type-dependent activity against the proteasome [24,25,29,48,54]. In one study, however, nelfinavir inhibited partially inhibited proteasome activity in breast cancer cell lysates, but caused ER stress differently from proteasome inhibitors [29]. Rather, the authors report evidence that the principal target of nelfinavir in these cells appeared to be a chaperone, heat shock protein 90, leading to ER stress as well as disruption of Her2 and Akt signaling. ER stress and cell killing can be increased by the combination of a PI (nelfinavir or ritonavir) and the proteasome inhibitor bortezomib in multiple cancer types (including those resistant to bortezomib alone) based on in vitro and mouse models [24,25,32,39,68,69]. ER stress and the UPR lead to autophagy, which results in cell survival if re-equilibration can be established, or in apoptosis and cell death in cases of overwhelming ER stress.
Autophagy
Autophagy is a catabolic process in which proteins and organelles are degraded and recycled either as a normal part of homeostasis, or in order to survive a period of nutrient starvation (reviewed in [70]). In addition to nutrient starvation, autophagy can be triggered by ER stress/UPR or PI3K/Akt inhibition caused by PIs or cancer chemotherapeutic agents [40]. Conversely, interfering with autophagy can cause ER stress and lead to cell death. As such, autophagy may protect cancer cells against death, and strategies that antagonize autophagy may complement the activity of therapies that induce ER stress/UPR. For example, an inhibitor of autophagy (3-methyladenine) increased nelfinavir-induced cell death in a panel of cancer cell lines [22]. In triple-negative breast cancer cell lines, the cytotoxic effects of ER stress induced by nelfinavir and/or celecoxib were synergistically enhanced by chloroquine, an inhibitor of autophagy [41]. Nelfinavir caused ER stress and autophagy in primary chronic lymphocytic leukemia cells from 14 patients; however, cytotoxicity was not observed unless nelfinavir was combined with chloroquine [42]. Apoptosis of prostate cancer cells treated with nelfinavir was also additively increased by hydroxycholorquine [43].
Apoptosis
PIs induce cell death through apoptosis in a variety of different cell lines [21,33,34,36,44-46,53,54], mediated either through ER stress or by other mechanisms, including inhibition of signaling through Akt, STAT3, c-Src, and NFαB [21,49]. PIs may inhibit NFαB activation through inhibition of proteasomal degradation of IαB [44,46,49,51,52]. Again, effects appear to be cell type-dependent; for example, interestingly, ritonavir may be protective against apoptosis in normal primary T cells [71].
Effects of nelfinavir on oncogenic herpesviruses
Nelfinavir, but not other PIs, is a potent inhibitor of Kaposi sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) replication in vitro with an EC50 ≤10 uM [72]. Nelfinavir does not act on the herpes protease (unpublished data); rather, its cellular effects appear to interfere with production of infectious KHSV. Given the importance of KSHV replication for KS pathogenesis, it is possible that nelfinavir could confer benefit through its antiviral as well as its antitumor properties [73]. Interestingly, nelfinavir also appears to trigger reactivation of KSHV and EBV in latently infected lymphoma cell lines, through induction of ER stress [69,74]. Nelfinavir’s effects on viral reactivation were independent of cytotoxicity. However, viral reactivation induced by nelfinavir in tumors might result in increased immune destruction and/or activity of nucleoside analog antivirals. Alternatively, increased expression of early viral genes like KSHV vIL-6 could have adverse consequences [75].
Clinical oncology trials (Table 2)
Table 2. Clinical oncology trials of HIV protease inhibitors.
Trial | PI and maximum dose |
PI-related toxicities | Reference |
---|---|---|---|
Endemic (HIV-negative Kaposi sarcoma) |
Indinavir, 800 mg BID |
Occasional modest skin and renal adverse effects reported |
[76] |
Pancreatic cancer (concomitant chemoradiotherapy with cisplatin, gemcitabine and radiation) |
Nelfinavir, 1250 mg BID |
Many toxicities associated with concomitant chemoradiation therapy, none attributed specifically to nelfinavir |
[77] |
Lung cancer (concomitant chemoradiotherapy with cis platin, etoposide and radiation) |
Nelfinavir, 1250 mg BID |
Many toxicities associated with concomitant chemoradiation therapy, none attributed specifically to nelfinavir |
[78] |
Refractory solid tumors | Nelfinavir, 3125 mg BID, dose escalation ongoing |
ALT, AST transaminitis, diarrhea, and hyperglycemia. None considered dose limiting. |
[79] |
Liposarcoma | Nelfinavir, 4250 mg BID |
1 grade 3 pancreatitis (reversed within 3 days), no other grade 3 or 4 toxicities; diarrhea, elevated AST, ALT; elevated glucose, elevated triglycerides |
[80] |
Based on their pre-clinical findings, Monini et al conducted a pilot trial of indinavir for endemic (HIV-negative) Kaposi sarcoma [76]. Twenty-eight patients received 800 mg of indinavir BID for 12 months, and although a control group was not included, clinical response was associated with blood levels of indinavir.
Two phase I studies of nelfinavir combined with chemoradiotherapy have been performed (Table 2). In patients with pancreatic cancer, nelfinavir 1250 mg BID was added to gemcitabine, cisplatin, and radiation [77] for its Akt inhibitory and radiosensitization properties. The authors reported that none of the observed toxicities appeared attributable to nelfinavir. Dose escalation was not pursued, because Akt phosphorylation in leukocytes was already reliably achieved at the standard nelfinavir dose. In patients with unresectable non-small-cell lung cancer, nelfinavir was studied at 625 mg BID or 1250 mg BID, in combination with cisplatin, etoposide, and radiation [78]. Again, no dose-limiting toxicities were observed and no toxicities were attributed to nelfinavir. The authors concluded that 1250 mg BID was an appropriate dose for further study with concurrent radiation chemotherapy.
In two phase I dose-escalation trials, nelfinavir has been studied as a single agent (Table 2). Among 11 evaluable patients with refractory solid tumors, nelfinavir was increased from 1250 mg BID to 3125 mg BID until the maximum tolerated dose was reached or there was disease progression [79]. No grade 4 or 5 toxicities were observed, and assays of peripheral blood mononuclear cells generally showed inhibition of Akt activation and increased markers of ER stress and apoptosis at every dose level. In patients with recurrent liposarcomas not amenable to surgical therapies, nelfinavir was administered orally BID for a 28 days/cycle in a phase 1/2 design [80]. Grade 3 pancreatitis requiring hospitalization was encountered at the lowest dose level, and reversed within 3 days; none of the pre-specified dose-limited toxicities were observed. Of note, pharmacokinetic data suggest that there may be only minimal increases in plasma drug concentrations with nelfinavir doses above 1875 mg BID, and that the drug induces its own clearance at doses of 4500 mg BID.
In sum, these trials show that nelfinavir: i) inhibits Akt-phosphorylation in blood leukocytes at standard doses in patients with and without cancer, ii) appears to be well tolerated even when administered with cytotoxic chemotherapy agents and concomitant radiation therapy, iii) may be administered as a single agent at higher than standard doses with acceptable toxicity, and iv) plasma levels may plateau with increasing doses and even decrease at very high doses. These observations and the preclinical data described above have motivated several ongoing clinical trials, including several phase 1/2 studies of patients with brain tumors, cervical cancer, pancreatic cancer, myeloma, lung cancer, and KS.
Conclusions
Epidemiologic and clinical studies of HIV-associated malignancies has led to recognition of the potential role for PIs in the treatment and prevention of a wide range of cancers. While as a class PIs may act similarly against HIV, they appear to be more heterogeneous in their effects on specific cellular pathways related to oncogenesis and inhibition of non-HIV viral production. Nelfinavir appears to be the most promising PI for the prevention and treatment of cancer, and a number of small clinical trials have begun to be conducted, showing encouraging tolerability and activity of nelfinavir-containing chemotherapeutic regimens for several cancer types in HIV-negative patients. Nelfinavir is highly attractive as a new cancer drug given that it is orally bioavailable and has an excellent safety profile, with more than 15 years of experience in of wide use in patients with HIV infection (including adults, children, and pregnant women, in resource-rich and resource-limited regions). These characteristics raise the possibility of novel chemopreventative strategies using nelfinavir in selected high risk settings, such as prevention of viral-associated malignancies, prevention of various cancers in chronically immunosuppressed patients, and secondary prevention of relapses.
Future studies should continue to define the differential antitumor effects among the different PIs, the basis for nelfinavir’s apparently superior activity, and the degree to which this varies by cell type. Such information would further the selection of the most synergistic combinations of chemotherapeutic drugs and the most appropriate tumors to target. In addition, an increased understanding of the PIs’ mechanisms of actions could lead to the development of PI analogs or derivative agents with greater potency, and result in more effective treatments for a wide range of cancers and viral infections.
Key Points.
PIs have been shown to kill a wide range of cancer cells types and inhibit angiogenesis in a large number of preclinical studies.
The mechanisms of action for these antitumor properties appear numerous, and include inhibition of Akt signaling, MMPs, and the proteasome, and induction of ER stress, autophagy, and apoptosis.
Nelfinavir appears to be the have the most broad and potent antitumor properties among the PIs, and has also been shown to have unique effects on oncogenic herpesvirus replication in vitro.
Nelfinavir is orally bioavailable, and has been shown through extensive use to be safe, and could be rapidly and efficiently repositioned as a cancer chemotherapeutic drug.
Early clinical studies are promising, but controlled trials are needed to evaluate the efficacy of nelfinavir against cancers in HIV-infected as well as uninfected patients.
Acknowledgements
Funding support: Support was provided from the following NIH Grants: P30 AI027757 (University of Washington Center for AIDS Research); U01 CA121947; R01 CA138165.
Footnotes
For all authors no conflicts of interest were declared.
References
- 1.Bernstein WB, Dennis PA. Repositioning HIV protease inhibitors as cancer therapeutics. Curr Opin HIV AIDS. 2008;3:666–675. doi: 10.1097/COH.0b013e328313915d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chow WA, Jiang C, Guan M. Anti-HIV drugs for cancer therapeutics: back to the future? Lancet Oncol. 2009;10:61–71. doi: 10.1016/S1470-2045(08)70334-6. [DOI] [PubMed] [Google Scholar]
- 3.Agouron Pharmaceuticals I . Viracept package insert. 2009. Edited by. [Google Scholar]
- 4.Adolescents PoAGfAa . In: Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents. Services DoHaH, editor. 2013. [Google Scholar]
- 5.Blum L, Pellet C, Agbalika F, Blanchard G, Morel P, Calvo F, Lebbe C. Complete remission of AIDS-related Kaposi’s sarcoma associated with undetectable human herpesvirus-8 sequences during anti-HIV protease therapy [letter] Aids. 1997;11:1653–1655. [PubMed] [Google Scholar]
- 6.Burdick AE, Carmichael C, Rady PL, Tyring SK, Badiavas E. Resolution of Kaposi’s sarcoma associated with undetectable level of human herpesvirus 8 DNA in a patient with AIDS after protease inhibitor therapy. J Am Acad Dermatol. 1997;37:648–649. doi: 10.1016/s0190-9622(97)70188-9. [DOI] [PubMed] [Google Scholar]
- 7.Conant MA, Opp KM, Poretz D, Mills RG. Reduction of Kaposi’s sarcoma lesions following treatment of AIDS with ritonovir. Aids. 1997;11:1300–1301. doi: 10.1097/00002030-199710001-00007. [DOI] [PubMed] [Google Scholar]
- 8.Murphy M, Armstrong D, Sepkowitz KA, Ahkami RN, Myskowski PL. Regression of AIDS-related Kaposi’s sarcoma following treatment with an HIV-1 protease inhibitor. Aids. 1997;11:261–262. [PubMed] [Google Scholar]
- 9.Diz Dios P, Ocampo Hermida A, Miralles Alvarez C, Vazquez Garcia E, Martinez Vazquez C. Regression of AIDS-related Kaposi’s sarcoma following ritonavir therapy. Oral Oncol. 1998;34:236–238. doi: 10.1016/s1368-8375(97)00082-1. [DOI] [PubMed] [Google Scholar]
- 10.Jung C, Bogner JR, Goebel F. Resolution of severe Kaposi’s sarcoma after initiation of antiretroviral triple therapy. Eur J Med Res. 1998;3:439–442. [PubMed] [Google Scholar]
- 11.Krischer J, Rutschmann O, Hirschel B, Vollenweider-Roten S, Saurat JH, Pechere M. Regression of Kaposi’s sarcoma during therapy with HIV-1 protease inhibitors: a prospective pilot study. J Am Acad Dermatol. 1998;38:594–598. doi: 10.1016/s0190-9622(98)70124-0. [DOI] [PubMed] [Google Scholar]
- 12.Lebbe C, Blum L, Pellet C, Blanchard G, Verola O, Morel P, Danne O, Calvo F. Clinical and biological impact of antiretroviral therapy with protease inhibitors on HIV-related Kaposi’s sarcoma. Aids. 1998;12:F45–49. doi: 10.1097/00002030-199807000-00002. [DOI] [PubMed] [Google Scholar]
- 13.De Milito A, Catucci M, Venturi G, Romano L, Incandela L, Valensin PE, Zazzi M. Antiretroviral therapy with protease inhibitors in human immunodeficiency virus type 1- and human herpesvirus 8-coinfected patients. J Med Virol. 1999;57:140–144. doi: 10.1002/(sici)1096-9071(199902)57:2<140::aid-jmv9>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 14.Gill J, Bourboulia D, Wilkinson J, Hayes P, Cope A, Marcelin AG, Calvez V, Gotch F, Boshoff C, Gazzard B. Prospective study of the effects of antiretroviral therapy on kaposi sarcoma-associated herpesvirus infection in patients with and without kaposi sarcoma. J Acquir Immune Defic Syndr. 2002;31:384–390. doi: 10.1097/00126334-200212010-00003. [DOI] [PubMed] [Google Scholar]
- 15.Martinez V, Caumes E, Gambotti L, Ittah H, Morini JP, Deleuze J, Gorin I, Katlama C, Bricaire F, Dupin N. Remission from Kaposi’s sarcoma on HAART is associated with suppression of HIV replication and is independent of protease inhibitor therapy. Br J Cancer. 2006;94:1000–1006. doi: 10.1038/sj.bjc.6603056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bower M, Weir J, Francis N, Newsom-Davis T, Powles S, Crook T, Boffito M, Gazzard B, Nelson M. The effect of HAART in 254 consecutive patients with AIDS-related Kaposi’s sarcoma. Aids. 2009;23:1701–1706. doi: 10.1097/QAD.0b013e32832d080d. [DOI] [PubMed] [Google Scholar]
- 17.Portsmouth S, Stebbing J, Gill J, Mandalia S, Bower M, Nelson M, Gazzard B. A comparison of regimens based on non-nucleoside reverse transcriptase inhibitors or protease inhibitors in preventing Kaposi’s sarcoma. Aids. 2003;17:F17–22. doi: 10.1097/00002030-200307250-00001. [DOI] [PubMed] [Google Scholar]
- 18.Grabar S, Abraham B, Mahamat A, Del Giudice P, Rosenthal E, Costagliola D. Differential Impact of Combination Antiretroviral Therapy in Preventing Kaposi’s Sarcoma With and Without Visceral Involvement. J Clin Oncol. 2006;24:3408–3414. doi: 10.1200/JCO.2005.05.4072. [DOI] [PubMed] [Google Scholar]
- 19.Crum-Cianflone NF, Hullsiek KH, Marconi V, Weintrob A, Ganesan A, Barthel RV, Fraser S, Roediger MP, Agan B, Wegner S. The impact of nelfinavir exposure on cancer development among a large cohort of HIV-infected patients. J Acquir Immune Defic Syndr. 2009;51:305–309. doi: 10.1097/QAI.0b013e3181aa13c7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gupta AK, Cerniglia GJ, Mick R, McKenna WG, Muschel RJ. HIV protease inhibitors block Akt signaling and radiosensitize tumor cells both in vitro and in vivo. Cancer Res. 2005;65:8256–8265. doi: 10.1158/0008-5472.CAN-05-1220. [DOI] [PubMed] [Google Scholar]
- 21.Yang Y, Ikezoe T, Takeuchi T, Adachi Y, Ohtsuki Y, Takeuchi S, Koeffler HP, Taguchi H. HIV-1 protease inhibitor induces growth arrest and apoptosis of human prostate cancer LNCaP cells in vitro and in vivo in conjunction with blockade of androgen receptor STAT3 and AKT signaling. Cancer Sci. 2005;96:425–433. doi: 10.1111/j.1349-7006.2005.00063.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gills JJ, Lopiccolo J, Tsurutani J, Shoemaker RH, Best CJ, Abu-Asab MS, Borojerdi J, Warfel NA, Gardner ER, Danish M, et al. Nelfinavir, A lead HIV protease inhibitor, is a broad-spectrum, anticancer agent that induces endoplasmic reticulum stress, autophagy, and apoptosis in vitro and in vivo. Clin Cancer Res. 2007;13:5183–5194. doi: 10.1158/1078-0432.CCR-07-0161. [DOI] [PubMed] [Google Scholar]
- 23.Zeng J, See AP, Aziz K, Thiyagarajan S, Salih T, Gajula RP, Armour M, Phallen J, Terezakis S, Kleinberg L, et al. Nelfinavir induces radiation sensitization in pituitary adenoma cells. Cancer biology & therapy. 2011;12:657–663. doi: 10.4161/cbt.12.7.17172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bono C, Karlin L, Harel S, Mouly E, Labaume S, Galicier L, Apcher S, Sauvageon H, Fermand JP, Bories JC, et al. The human immunodeficiency virus-1 protease inhibitor nelfinavir impairs proteasome activity and inhibits the proliferation of multiple myeloma cells in vitro and in vivo. Haematologica. 2012;97:1101–1109. doi: 10.3324/haematol.2011.049981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kraus M, Bader J, Overkleeft H, Driessen C. Nelfinavir augments proteasome inhibition by bortezomib in myeloma cells and overcomes bortezomib and carfilzomib resistance. Blood cancer journal. 2013;3:e103. doi: 10.1038/bcj.2013.2. ** A careful study showing that in multiple myeloma cells, nelfinavir had the greatest cytotoxic activity among the PIs, and displayed synergy with bortezomib/carfilzomib even in bortezomib/carfilzomib resistant cells. This effect appeared to be due to nelfinavir’s greater activity on the proteasome inhibition or ER stress, rather than Akt inhibition, which was comparable among all nine PIs tested.
- 26.Yang Y, Ikezoe T, Nishioka C, Bandobashi K, Takeuchi T, Adachi Y, Kobayashi M, Takeuchi S, Koeffler HP, Taguchi H. NFV, an HIV-1 protease inhibitor, induces growth arrest, reduced Akt signalling, apoptosis and docetaxel sensitisation in NSCLC cell lines. Br J Cancer. 2006;95:1653–1662. doi: 10.1038/sj.bjc.6603435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pore N, Gupta AK, Cerniglia GJ, Jiang Z, Bernhard EJ, Evans SM, Koch CJ, Hahn SM, Maity A. Nelfinavir down-regulates hypoxia-inducible factor 1alpha and VEGF expression and increases tumor oxygenation: implications for radiotherapy. Cancer Res. 2006;66:9252–9259. doi: 10.1158/0008-5472.CAN-06-1239. [DOI] [PubMed] [Google Scholar]
- 28.Jiang Z, Pore N, Cerniglia GJ, Mick R, Georgescu MM, Bernhard EJ, Hahn SM, Gupta AK, Maity A. Phosphatase and tensin homologue deficiency in glioblastoma confers resistance to radiation and temozolomide that is reversed by the protease inhibitor nelfinavir. Cancer Res. 2007;67:4467–4473. doi: 10.1158/0008-5472.CAN-06-3398. [DOI] [PubMed] [Google Scholar]
- 29.Shim JS, Rao R, Beebe K, Neckers L, Han I, Nahta R, Liu JO. Selective inhibition of HER2-positive breast cancer cells by the HIV protease inhibitor nelfinavir. Journal of the National Cancer Institute. 2012;104:1576–1590. doi: 10.1093/jnci/djs396. ** A detailed study describing the selective growth inhibiton of Her2-positive breast cancer cells by nelfinavir in vitro, and evidence that nelfinavir acts on HSP90, rather than the proteasome.
- 30.Petrich AM, Leshchenko V, Kuo PY, Xia B, Thirukonda VK, Ulahannan N, Gordon S, Fazzari MJ, Ye BH, Sparano JA, et al. Akt inhibitors MK-2206 and nelfinavir overcome mTOR inhibitor resistance in diffuse large B-cell lymphoma. Clinical cancer research: an official journal of the American Association for Cancer Research. 2012;18:2534–2544. doi: 10.1158/1078-0432.CCR-11-1407. ** A panel of DLBCL cell lines was studied using pathway analysis and connectivity mapping, which identified Akt as central to the rapamycin resistance. Nelfinavir and MK-2206, both inhibitors of Akt, were synergistic with rapamycin in killling DLBCL cell lines.
- 31.Gupta AK, Li B, Cerniglia GJ, Ahmed MS, Hahn SM, Maity A. The HIV protease inhibitor nelfinavir downregulates Akt phosphorylation by inhibiting proteasomal activity and inducing the unfolded protein response. Neoplasia. 2007;9:271–278. doi: 10.1593/neo.07124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sato A, Asano T, Ito K. Ritonavir interacts with bortezomib to enhance protein ubiquitination and histone acetylation synergistically in renal cancer cells. Urology. 2012;79:966, e913–921. doi: 10.1016/j.urology.2011.11.033. [DOI] [PubMed] [Google Scholar]
- 33.Bruning A, Vogel M, Mylonas I, Friese K, Burges A. Bortezomib targets the caspase-like proteasome activity in cervical cancer cells, triggering apoptosis that can be enhanced by nelfinavir. Current cancer drug targets. 2011;11:799–809. doi: 10.2174/156800911796798913. [DOI] [PubMed] [Google Scholar]
- 34.Tian X, Ye J, Alonso-Basanta M, Hahn SM, Koumenis C, Dorsey JF. Modulation of CCAAT/enhancer binding protein homologous protein (CHOP)-dependent DR5 expression by nelfinavir sensitizes glioblastoma multiforme cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) The Journal of biological chemistry. 2011;286:29408–29416. doi: 10.1074/jbc.M110.197665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Pyrko P, Kardosh A, Wang W, Xiong W, Schonthal AH, Chen TC. HIV-1 protease inhibitors nelfinavir and atazanavir induce malignant glioma death by triggering endoplasmic reticulum stress. Cancer Res. 2007;67:10920–10928. doi: 10.1158/0008-5472.CAN-07-0796. [DOI] [PubMed] [Google Scholar]
- 36.Bruning A, Burger P, Vogel M, Rahmeh M, Gingelmaiers A, Friese K, Lenhard M, Burges A. Nelfinavir induces the unfolded protein response in ovarian cancer cells, resulting in ER vacuolization, cell cycle retardation and apoptosis. Cancer Biol Ther. 2009;8 doi: 10.4161/cbt.8.3.7339. [DOI] [PubMed] [Google Scholar]
- 37.Bruning A. Analysis of nelfinavir-induced endoplasmic reticulum stress. Methods in enzymology. 2011;491:127–142. doi: 10.1016/B978-0-12-385928-0.00008-0. [DOI] [PubMed] [Google Scholar]
- 38.Guan M, Fousek K, Jiang C, Guo S, Synold T, Xi B, Shih CC, Chow WA. Nelfinavir induces liposarcoma apoptosis through inhibition of regulated intramembrane proteolysis of SREBP-1 and ATF6. Clinical cancer research: an official journal of the American Association for Cancer Research. 2011;17:1796–1806. doi: 10.1158/1078-0432.CCR-10-3216. [DOI] [PubMed] [Google Scholar]
- 39.Kawabata S, Gills JJ, Mercado-Matos JR, Lopiccolo J, Wilson W, 3rd, Hollander MC, Dennis PA. Synergistic effects of nelfinavir and bortezomib on proteotoxic death of NSCLC and multiple myeloma cells. Cell death & disease. 2012;3:e353. doi: 10.1038/cddis.2012.87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gills JJ, Lopiccolo J, Dennis PA. Nelfinavir, a new anti-cancer drug with pleiotropic effects and many paths to autophagy. Autophagy. 2008;4:107–109. doi: 10.4161/auto.5224. [DOI] [PubMed] [Google Scholar]
- 41.Thomas S, Sharma N, Golden EB, Cho H, Agarwal P, Gaffney KJ, Petasis NA, Chen TC, Hofman FM, Louie SG, et al. Preferential killing of triple-negative breast cancer cells in vitro and in vivo when pharmacological aggravators of endoplasmic reticulum stress are combined with autophagy inhibitors. Cancer letters. 2012;325:63–71. doi: 10.1016/j.canlet.2012.05.030. [DOI] [PubMed] [Google Scholar]
- 42.Mahoney E, Maddocks K, Flynn J, Jones J, Cole SL, Zhang X, Byrd JC, Johnson AJ. Identification Of Endoplasmic Reticulum Stress Inducing Agents By Antagonizing Autophagy: A New Potential Strategy For Identification Of Anti-Cancer Therapeutics In B-Cell Malignancies. Leukemia & lymphoma. 2013 doi: 10.3109/10428194.2013.781168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Guan M, Fousek K, Chow WA. Nelfinavir inhibits regulated intramembrane proteolysis of sterol regulatory element binding protein-1 and activating transcription factor 6 in castration-resistant prostate cancer. The FEBS journal. 2012;279:2399–2411. doi: 10.1111/j.1742-4658.2012.08619.x. [DOI] [PubMed] [Google Scholar]
- 44.Pati S, Pelser CB, Dufraine J, Bryant JL, Reitz MS, Jr., Weichold FF. Antitumorigenic effects of HIV protease inhibitor ritonavir: inhibition of Kaposi sarcoma. Blood. 2002;99:3771–3779. doi: 10.1182/blood.v99.10.3771. [DOI] [PubMed] [Google Scholar]
- 45.Sun L, Niu L, Zhu X, Hao J, Wang P, Wang H. Antitumour effects of a protease inhibitor, nelfinavir, in hepatocellular carcinoma cancer cells. Journal of chemotherapy. 2012;24:161–166. doi: 10.1179/1973947812Y.0000000011. [DOI] [PubMed] [Google Scholar]
- 46.Dewan MZ, Uchihara JN, Terashima K, Honda M, Sata T, Ito M, Fujii N, Uozumi K, Tsukasaki K, Tomonaga M, et al. Efficient intervention of growth and infiltration of primary adult T-cell leukemia cells by an HIV protease inhibitor, ritonavir. Blood. 2006;107:716–724. doi: 10.1182/blood-2005-02-0735. [DOI] [PubMed] [Google Scholar]
- 47.Hampson L, Kitchener HC, Hampson IN. Specific HIV protease inhibitors inhibit the ability of HPV16 E6 to degrade p53 and selectively kill E6-dependent cervical carcinoma cells in vitro. Antivir Ther. 2006;11:813–825. [PubMed] [Google Scholar]
- 48.Jiang W, Mikochik PJ, Ra JH, Lei H, Flaherty KT, Winkler JD, Spitz FR. HIV protease inhibitor nelfinavir inhibits growth of human melanoma cells by induction of cell cycle arrest. Cancer Res. 2007;67:1221–1227. doi: 10.1158/0008-5472.CAN-06-3377. [DOI] [PubMed] [Google Scholar]
- 49.Dewan MZ, Tomita M, Katano H, Yamamoto N, Ahmed S, Yamamoto M, Sata T, Mori N. An HIV protease inhibitor, ritonavir targets the nuclear factor-kappaB and inhibits the tumor growth and infiltration of EBV-positive lymphoblastoid B cells. International journal of cancer. Journal international du cancer. 2009;124:622–629. doi: 10.1002/ijc.23993. [DOI] [PubMed] [Google Scholar]
- 50.Pajonk F, Himmelsbach J, Riess K, Sommer A, McBride WH. The human immunodeficiency virus (HIV)-1 protease inhibitor saquinavir inhibits proteasome function and causes apoptosis and radiosensitization in non-HIV-associated human cancer cells. Cancer research. 2002;62:5230–5235. [PubMed] [Google Scholar]
- 51.Srirangam A, Milani M, Mitra R, Guo Z, Rodriguez M, Kathuria H, Fukuda S, Rizzardi A, Schmechel S, Skalnik DG, et al. The human immunodeficiency virus protease inhibitor ritonavir inhibits lung cancer cells, in part, by inhibition of survivin. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer. 2011;6:661–670. doi: 10.1097/JTO.0b013e31820c9e3c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Timeus F, Crescenzio N, Doria A, Foglia L, Pagliano S, Ricotti E, Fagioli F, Tovo PA, Cordero di Montezemolo L. In vitro anti-neuroblastoma activity of saquinavir and its association with imatinib. Oncology reports. 2012;27:734–740. doi: 10.3892/or.2011.1582. * A paper demonstrating that saquinavir has a dose-dependent anti-proliferative and anti-invasive activity on NB lines, that was potentiated by imatinib, with both at clinically attainable concentrations. These effects appeared to be due to inhibition of NF-kappaB and possibly induction of apoptosis.
- 53.Srirangam A, Mitra R, Wang M, Gorski JC, Badve S, Baldridge L, Hamilton J, Kishimoto H, Hawes J, Li L, et al. Effects of HIV protease inhibitor ritonavir on Akt-regulated cell proliferation in breast cancer. Clinical cancer research: an official journal of the American Association for Cancer Research. 2006;12:1883–1896. doi: 10.1158/1078-0432.CCR-05-1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gaedicke S, Firat-Geier E, Constantiniu O, Lucchiari-Hartz M, Freudenberg M, Galanos C, Niedermann G. Antitumor effect of the human immunodeficiency virus protease inhibitor ritonavir: induction of tumor-cell apoptosis associated with perturbation of proteasomal proteolysis. Cancer research. 2002;62:6901–6908. [PubMed] [Google Scholar]
- 55.Barillari G, Iovane A, Bacigalupo I, Palladino C, Bellino S, Leone P, Monini P, Ensoli B. Ritonavir or saquinavir impairs the invasion of cervical intraepithelial neoplasia cells via a reduction of MMP expression and activity. AIDS. 2012;26:909–919. doi: 10.1097/QAD.0b013e328351f7a5. [DOI] [PubMed] [Google Scholar]
- 56.Esposito V, Verdina A, Manente L, Spugnini EP, Viglietti R, Parrella R, Pagliano P, Parrella G, Galati R, De Luca A, et al. Amprenavir inhibits the migration in human hepatocarcinoma cell and the growth of xenografts. Journal of cellular physiology. 2013;228:640–645. doi: 10.1002/jcp.24173. [DOI] [PubMed] [Google Scholar]
- 57.Sgadari C, Barillari G, Toschi E, Carlei D, Bacigalupo I, Baccarini S, Palladino C, Leone P, Bugarini R, Malavasi L, et al. HIV protease inhibitors are potent anti-angiogenic molecules and promote regression of Kaposi sarcoma. Nat Med. 2002;8:225–232. doi: 10.1038/nm0302-225. [DOI] [PubMed] [Google Scholar]
- 58.Andre P, Groettrup M, Klenerman P, de Giuli R, Booth BL, Jr., Cerundolo V, Bonneville M, Jotereau F, Zinkernagel RM, Lotteau V. An inhibitor of HIV-1 protease modulates proteasome activity, antigen presentation, and T cell responses. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:13120–13124. doi: 10.1073/pnas.95.22.13120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Xie L, Evangelidis T, Bourne PE. Drug discovery using chemical systems biology: weak inhibition of multiple kinases may contribute to the anti-cancer effect of nelfinavir. PLoS computational biology. 2011;7:e1002037. doi: 10.1371/journal.pcbi.1002037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lucia MB, Anu R, Handley M, Gillet JP, Wu CP, De Donatis GM, Cauda R, Gottesman MM. Exposure to HIV-protease inhibitors selects for increased expression of P-glycoprotein (ABCB1) in Kaposi’s sarcoma cells. British journal of cancer. 2011;105:513–522. doi: 10.1038/bjc.2011.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Pore N, Gupta AK, Cerniglia GJ, Maity A. HIV protease inhibitors decrease VEGF/HIF-1alpha expression and angiogenesis in glioblastoma cells. Neoplasia. 2006;8:889–895. doi: 10.1593/neo.06535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Bartholomeusz C, Gonzalez-Angulo AM. Targeting the PI3K signaling pathway in cancer therapy. Expert opinion on therapeutic targets. 2012;16:121–130. doi: 10.1517/14728222.2011.644788. [DOI] [PubMed] [Google Scholar]
- 63.Burris HA., 3rd Overcoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathway. Cancer chemotherapy and pharmacology. 2013 doi: 10.1007/s00280-012-2043-3. [DOI] [PubMed] [Google Scholar]
- 64.Plastaras JP, Vapiwala N, Ahmed MS, Gudonis D, Cerniglia GJ, Feldman MD, Frank I, Gupta AK. Validation and toxicity of PI3K/Akt pathway inhibition by HIV protease inhibitors in humans. Cancer Biol Ther. 2008;7:628–635. doi: 10.4161/cbt.7.5.5728. [DOI] [PubMed] [Google Scholar]
- 65.Schonthal AH. Pharmacological targeting of endoplasmic reticulum stress signaling in cancer. Biochemical pharmacology. 2013;85:653–666. doi: 10.1016/j.bcp.2012.09.012. [DOI] [PubMed] [Google Scholar]
- 66.Martinon F. Targeting endoplasmic reticulum signaling pathways in cancer. Acta oncologica. 2012;51:822–830. doi: 10.3109/0284186X.2012.689113. [DOI] [PubMed] [Google Scholar]
- 67.Chow WA, Guo S, Valdes-Albini F. Nelfinavir induces liposarcoma apoptosis and cell cycle arrest by upregulating sterol regulatory element binding protein-1. Anticancer Drugs. 2006;17:891–903. doi: 10.1097/01.cad.0000224448.08706.76. [DOI] [PubMed] [Google Scholar]
- 68.Kraus M, Malenke E, Gogel J, Muller H, Ruckrich T, Overkleeft H, Ovaa H, Koscielniak E, Hartmann JT, Driessen C. Ritonavir induces endoplasmic reticulum stress and sensitizes sarcoma cells toward bortezomib-induced apoptosis. Molecular cancer therapeutics. 2008;7:1940–1948. doi: 10.1158/1535-7163.MCT-07-2375. [DOI] [PubMed] [Google Scholar]
- 69.Shirley C, Kalu N, Shamay M, Ambinder RF. Epstein-Barr virus lytic gene expression is tightly linked to ER stress but not cytotoxicity with bortezomib or nelfinavir; International Congress on Oncogenic Herpesviruses and Associated Diseases; Philadelphia, PA: Infectious Agents and Cancer. 2012. p. P32. [Google Scholar]
- 70.Choi AM, Ryter SW, Levine B. Autophagy in human health and disease. The New England journal of medicine. 2013;368:651–662. doi: 10.1056/NEJMra1205406. [DOI] [PubMed] [Google Scholar]
- 71.Weichold FF, Bryant JL, Pati S, Barabitskaya O, Gallo RC, Reitz MS., Jr. HIV-1 protease inhibitor ritonavir modulates susceptibility to apoptosis of uninfected T cells. Journal of human virology. 1999;2:261–269. [PubMed] [Google Scholar]
- 72.Gantt S, Carlsson J, Ikoma M, Gachelet E, Gray M, Geballe AP, Corey L, Casper C, Lagunoff M, Vieira J. The HIV protease inhibitor nelfinavir inhibits Kaposi’s sarcoma-associated herpesvirus replication in vitro. Antimicrobial agents and chemotherapy. 2011;55:2696–2703. doi: 10.1128/AAC.01295-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gantt S, Casper C. Human herpesvirus 8-associated neoplasms: the roles of viral replication and antiviral treatment. Curr Opin Infect Dis. 2011;24:295–301. doi: 10.1097/QCO.0b013e3283486d04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kalu N, Shirley C, Ambinder RF. ER stress activates lytic gene expression in KSHV associated tumor cell lines; International Congress on Oncogenic Herpesviruses and Associated Diseases; Philadelphia, PA: Infectious Agents and Cancer. 2012.p. 33. [Google Scholar]
- 75.Ganem D. KSHV and the pathogenesis of Kaposi sarcoma: listening to human biology and medicine. J Clin Invest. 120:939–949. doi: 10.1172/JCI40567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Monini P, Sgadari C, Grosso MG, Bellino S, Di Biagio A, Toschi E, Bacigalupo I, Sabbatucci M, Cencioni G, Salvi E, et al. Clinical course of classic Kaposi’s sarcoma in HIV-negative patients treated with the HIV protease inhibitor indinavir. Aids. 2009;23:534–538. doi: 10.1097/QAD.0b013e3283262a8d. [DOI] [PubMed] [Google Scholar]
- 77.Brunner TB, Geiger M, Grabenbauer GG, Lang-Welzenbach M, Mantoni TS, Cavallaro A, Sauer R, Hohenberger W, McKenna WG. Phase I trial of the human immunodeficiency virus protease inhibitor nelfinavir and chemoradiation for locally advanced pancreatic cancer. J Clin Oncol. 2008;26:2699–2706. doi: 10.1200/JCO.2007.15.2355. [DOI] [PubMed] [Google Scholar]
- 78.Rengan R, Mick R, Pryma D, Rosen MA, Lin LL, Maity AM, Evans TL, Stevenson JP, Langer CJ, Kucharczuk J, et al. A phase I trial of the HIV protease inhibitor nelfinavir with concurrent chemoradiotherapy for unresectable stage IIIA/IIIB non-small cell lung cancer: a report of toxicities and clinical response. Journal of thoracic oncology: official publication of the International Association for the Study of Lung Cancer. 2012;7:709–715. doi: 10.1097/JTO.0b013e3182435aa6. * Nelfinavir was given in combination with cisplatin, etoposide, and radiation. There appeared to be no toxicities that were attributable to nelfinavir, suggesting that a dose of 1250 mg BID with concurrent radiation chemotherapy was appropriate for further study.
- 79.Dennis P, Blumenthal G, Ballas M, Gardner E, Kawabata S, LoPiccolo J, Helsabeck C, Root H, Figg W, Bernstein W. A phase I study of nelfinavir, an FDA approved HIV protease inhibitor, in adults with refractory solid tumors. J Clin Oncol (Meeting Abstracts) 2009:2583. [Google Scholar]
- 80.Pan J, Mott M, Xi B, Hepner E, Guan M, Fousek K, Magnusson R, Tinsley R, Valdes F, Frankel P, et al. Phase I study of nelfinavir in liposarcoma. Cancer Chemother Pharmacol. 2012;70:791–799. doi: 10.1007/s00280-012-1961-4. ** This is a dose-escalation study of nelfinavir for liposarcoma. No dose-limited toxicities were observed. Pharmacokinetic analyses suggest that there may be only minimal increases in plasma drug concentrations with nelfinavir doses above 1875 mg BID, and that the drug induces its own clearance at doses of 4500 mg BID.