Main

Until a few years ago, infection by the human immuno-deficiency virus (HIV) was inexorably leading to AIDS, characterized by immunodeficiency, opportunistic infections and increased incidence and aggressiveness of certain types of tumour, particularly Kaposi's sarcoma, B-cell lymphomas and anogenital cancer (Table 1)1. The advent of the highly active antiretroviral therapy (HAART), however, has turned HIV infection into a manageable disease2. HAART is a combination therapy against HIV that comprises several active molecules that block multiple viral targets (Box 1). At present, HAART uses drugs directed against the viral reverse transcriptase — nucleoside and non-nucleoside reverse-transcriptase inhibitors (NRTIs and NNRTIs, respectively) — and drugs directed against the viral aspartyl protease, known as HIV-protease inhibitors (HIV-PIs)3. NRTIs act by blocking the HIV reverse-transcriptase active site, whereas NNRTIs inhibit the activity of HIV reverse transcriptase by binding to other sites present on the enzyme (allosteric inhibition)3. HIV-PIs are peptide mimetic drugs that have been designed to mimic the phenylalanine–proline peptide bond, which is targeted by the viral protease, but is not cleaved by any of the known mammalian endopeptidases. This confers a remarkable specificity of action to HIV-PIs that, on short-term treatment, show only mild side effects and a tolerable toxicity. So, two types of HAART regimens are in use at present, and include a combination of traditional NRTIs with either NNRTIs (NNRTI–HAART) or HIV-PIs (PI–HAART) — these are similarly effective in suppressing HIV infection3.

Table 1 HIV-associated malignancies

The rationale for these combination therapies is to inhibit several steps of the viral life cycle. Combination regimens based on HIV-PIs or NNRTIs have been shown to be more effective than single or dual combinations of NRTIs in suppressing HIV replication, and in preserving or reconstituting both NAIVE AND MEMORY T-CELL REPERTOIRES, which delays or reverts the onset of AIDS4,5,6,7. HAART has also led to a lower incidence and/or regression of Kaposi's sarcoma, non-Hodgkin's lymphomas (NHLs) and intra-epithelial anogenital tumours8,9.

Antiviral drugs included in HAART regimens are known to exert a direct antitumour action. In particular, azidothymidine (AZT), one of the most widely used NRTIs, has been shown to block telomerase activity, to induce TRIAL-mediated apoptosis of lymphomatous cells and to reverses cisplatin resistance in patients with cancer10,11,12. AZT therapy also leads to a complete remission of advanced Kaposi's sarcoma in HIV-infected patients treated with chemotherapy13. Therapies based on single or dual NRTI regimens have also decreased the incidence of Kaposi's sarcoma and NHLs14,15,16. However, this reduction is much lower than the marked decrease in the incidence of these tumours in patients treated with HAART.

These antitumour effects of HAART regimens have been attributed to the extraordinary efficacy of this drug combination in suppressing HIV infection, and to the consequent reconstitution of cellular immunity against viruses implicated in AIDS-associated malignancies, including Epstein–Barr virus (EBV), Kaposi's-sarcoma-associated herpesvirus (KSHV) or human papillomavirus (HPV). This model, however, does not explain why these antiretroviral regimens did not show similar effects on other HIV-related proliferative diseases associated with the same viruses, including EPIDERMODYSPLASIA VERRUCIFORMS, MULTICENTRIC CASTLEMAN'S DISEASE, HODGKIN'S DISEASE or invasive cervical cancer8,17,18. Moreover, some HIV-infected patients who still experience persistent and profound immune deficiency despite HAART treatment still have a much lower risk of developing NHLs, compared with similarly immune-suppressed individuals who have been treated with single or dual combinations of NRTIs19. This indicates that HAART could have direct antitumour effects that are unrelated to the use of NRTIs and are independent of immune reconstitution.

There is much evidence that HIV-PIs have direct effects on angiogenesis, tumour growth and tumour immunity that are unrelated to their antiviral effects. These seem to be mediated through specific activities on the proteasome and on MATRIX METALLOPROTEINASES (MMPs)20,21,22.

HIV infection and tumour development

AIDS-associated malignancies are all associated with DNA tumour viruses such as EBV, KSHV and HPV (Table 1). For this reason, these malignancies are commonly considered to be the result of diminished immune surveillance against viruses and virus-infected tumour cells. The beneficial effects of HAART on these tumours have therefore been interpreted as the result of drug-mediated HIV suppression and immune reconstitution. This is supported by several findings. For example, Kaposi's sarcoma is a multifocal angiogenic tumour characterized by proliferating spindle-shaped cells of endothelial-cell origin that are latently infected by KSHV23. Studies indicate that KSHV load and antibody titres, as well as the patient's number of CD4+ HELPER T LYMPHOCYTES — the main targets of HIV replication24,25,26,32 — are independent determinants for risk of developing Kaposi's sarcoma25,26,27,28,29,30,31. Similarly, EBV load is increased in patients before development of B-cell lymphoma, whereas specific immune responses against the virus are decreased33,34,35. The relative risk of AIDS-associated malignancies increases progressively as a function of the progressive decline of CD4+ T-cell counts27.

Nevertheless, the relation between immune deficiency and tumour development is not straightforward. In fact, only certain types of AIDS-associated tumours arise in immunodeficient patients. In particular, NHL subtypes including IMMUNOBLASTIC LYMPHOMAS and PRIMARY-NERVOUS-SYSTEM LYMPHOMAS, along with Burkitt's-like lymphomas, typically develop in pa tients with very low CD4+ T-cell counts. On the other hand, the incidence of other NHL subtypes such as CENTROBLASTIC DIFFUSE LARGE-CELL LYMPHOMAS, along with classic BURKITT'S LYMPHOMA, Hodgkin's disease, cervical cancer and, most notably, Kaposi's sarcoma, increases in patients who have significantly higher CD4+ T-cell numbers1,19,36,37,38,39,40,41.

The overall risk of tumour development is very high in HIV-infected individuals (Table 1), but the relative increase in tumour risk with stepwise decreases in CD4+ T-cell counts is only marginal27. So the risk of tumour development increases steeply as CD4+ T-cell counts decline below a certain threshold. Once below this threshold, cancer risk becomes less dependent on further CD4+ T-cell loss27. However, evidence indicates that this hypothetical CD4+ T-cell count threshold can be very high in certain individuals. In particular, in HIV-infected homosexual men, the incidence rate of Kaposi's sarcoma increases by more than 1000-fold before a consistent CD4+ T-cell decline1. So, CD4+ T-cell loss and consequent immune deficiency cannot fully explain the increased incidence of certain malignancies in HIV-infected individuals. Indeed, several recent studies show that immune activation causes and precedes the development of immune deficiency in HIV infection42,43,44,45. Sustained and uncontrolled HIV replication leads to continuous antigenic stimulation and to chronic T-cell activation and proliferation, which, in turn, generates a continuous drain of naive and memory T cells that become activated, proliferate, die by apoptosis or re-enter the pool of memory T cells. However, this exhausts the pool of naive T cells, impairing the capacity to mount antigen-specific immune responses42,43,44,45 (Box 2).

Several other studies also indicate that immune activation, rather than immune deficiency, is the key factor in the initiation of Kaposi's sarcoma and B-cell lymphomas. In particular, Kaposi's sarcoma begins as a reactive process, characterized by blood-vessel activation (expression by endothelial cells of adhesion molecules and αvβ3- and α5β1-integrins), tissue infiltration by activated CD4+ and CD8+ T cells, production of angiogenic factors, increased expression of MMPs, and intense and aberrant angiogenesis23. Extracellular HIV-1 Tat PROTEIN, released by HIV-infected cells, binds the αvβ3- and α5β1-integrins, increasing the effects of the angiogenic factors46,47,48,49,50,51,52,53. These processes can precede the appearance of spindle-shaped cells of endothelial or monocytic cell origin (the so-called Kaposi's-sarcoma spindle cells), which are believed to be the tumour cells of Kaposi's sarcoma54,55 and becomes progressively infected by KSHV23.

Evidence indicates that all these events, including KSHV reactivation and virus dissemination to tissues, are driven by the actions of TH1-TYPE PRO-INFLAMMATORY CYTOKINES, particularly interferon-γ (IFNγ), interleukin-1β (IL-1β) and tumour-necrosis factor-α (TNFα), which are increased in patients infected by HIV23,46,48,55,56,57,58,59,60. This is also supported by the observation that treatment of HIV-infected patients with IL-2, TNFα, or IFNγ resulted in onset or progression of Kaposi's sarcoma61,62. Furthermore, a CD8/TH1-type immune-activation profile is a specific trait of individuals who are at risk of forms of Kaposi's sarcoma that are not associated with HIV infection, including CLASSIC KAPOSI'S SARCOMA23,63,64,65,66 and AFRICAN KAPOSI'S SARCOMA23,67,68,69. Moreover, TH1-type cytokines are also likely to be involved in the initiation of POST-TRANSPLANT KAPOSI'S SARCOMA. In fact, only a small fraction of KSHV-infected organ recipients develop Kaposi's sarcoma following initiation of immune-suppressive therapy70, whereas disease onset is most frequently preceded by acute or chronic graft rejection or opportunistic infections, which induce strong TH1-type responses71,72. AIDS-associated B-cell lymphomas are also preceded by chronic antigen-dependent B-cell stimulation leading to a persistent and generalized lymphadenopathy that, in turn, promotes the clonal expansion of pre-neoplastic antigen-specific B-cell populations73,74. Furthermore, an increased EBV load precedes the development of B-cell lymphoma34, whereas extracellular Tat increases B-cell proliferation and induces B-cell lymphomas in mice75,76.

So, AIDS-associated malignancies are multifactorial and their progression is promoted by the combined effects of immune activation, viral cofactors (including tumour viruses and extracellular Tat) and immune deficiency. In this regard, two variables are most often measured to monitor HIV infection and the efficacy of antiretroviral therapies — the number of CD4+ T cells, which is a measure of immune integrity or deficiency, and HIV load. In HAART-treated patients, HIV load remains high in non-responders or in patients showing persistent viraemia despite an increase in their CD4+ T-cell numbers. These individuals have high or intermediate levels of immune activation compared with patients who respond to HAART. So, HIV load might be regarded as an indirect measure of immune activation in treated patients. Recent data, however, have indicated that the effects of HAART on AIDS-associated tumours are not always well correlated to either one of these two key variables, so HAART might exert direct antitumour actions that act independently of immune reconstitution or suppression of immune activation.

HAART inhibits AIDS-associated tumours

Following HAART initiation, patients show a sudden decrease in HIV replication associated with a reduction of both T-cell and B-cell activation and turnover rates4,43,44,77,78,79,80,81. This leads to an increase in memory and naive CD4+ and CD8+ T-cell counts, normalization of the B-cell compartment and, eventually, sustained immune reconstitution4,78,80,82,83. These effects of HAART are associated with a significant decrease in the incidence of AIDS-associated malignancies — particularly in the incidence of Kaposi's sarcoma and NHLs — as well as with tumour regression, prolonged time to failure of tumour treatment and longer survival times8,84,85,86,87,88,89,90,91,92,93. These positive outcomes have been generally attributed to restoration of the immunological control of viruses, including EBV, KSHV and HPV19,84,94,95,96,97.

Studies comparing the incidence of lymphomas before or after the advent of HAART indicated that HAART resulted in a significant decrease in the incidence of systemic immunoblastic lymphomas and primary-nervous-system lymphomas, but not of the other lymphoma subtypes8,15,98,99. This has been generally attributed to drug-mediated reconstitution of immune responses against cells that express immunogenic EBV latency gene products, including the latency membrane protein 1 (LMP1) oncoprotein, which is prominently expressed by immunoblastic lymphomas and primary-nervous-system lymphomas (Table 1)74,100,101,102. However, recent retrospective studies that compared the incidence of lymphomas in groups of patients treated with HAART or with NRTIs, and patients who did not receive antiretroviral therapy showed that the incidence of all lymphoma subtypes is reduced in patients who have received HAART. So, HAART reduces the incidence of NHLs whether or not their pathogenesis depends on immune suppression19,95.

A high rate of tumour regression and/or complete remission is frequently observed in patients with Kaposi's sarcoma and NHL who have been treated with either chemotherapy or radiotherapy and HAART, or with HAART alone9,84,88,90,91,96,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117. Although larger, controlled studies are required, these data indicate that HAART not only reduces tumour incidence in HIV-infected individuals, but it also induces an antitumour response. However, although increased CD4+ T-cell counts and decreased or undetectable levels of HIV and KSHV viraemia are independent predictors of the response of Kaposi's sarcoma to HAART84,103,105,108,118, several studies reported a lack of correlation between one or more of these determinants and tumour regression107,119,120,121. A recent study has shown that HAART prolongs the length of time that patients with Kaposi's sarcoma respond to chemotherapy, but this effect is not well correlated with restoration of CD4+ T cells or with control of HIV viraemia87. In another recent study, reconstitution of immune responses against KSHV and decreases of plasma viraemic levels following HAART have been shown to require several months of therapy (over 24 months) — much longer than the rapid response of Kaposi's sarcoma to these drugs122. This tumour, in fact, typically responds to HAART within the first weeks of treatment, often resulting in complete disease remission after only a few months of therapy122.

Similarly, regression of cervical intraepithelial neoplasia occurs following HAART initiation despite persistence of HPV DNA in the cervix9,117. By contrast, in untreated patients, persistence of HPV infection leads to lesion progression123,124. Although regression of cervical intraepithelial neoplasia during HAART is associated with immune reconstitution, a lack of correlation between CD4+ T-cell counts, HIV load, clearance of HPV infection and lesion regression has been reported9,96,117. All these data confirm that regression or resolution of Kaposi's sarcoma, NHLs and cervical intraepithelial neoplasias in response to HAART are mediated, at least in part, by effects that are not due to suppression of HIV infection, inhibition of antigen-driven immune activation, or HAART-mediated immune reconstitution.

Antitumour effects of HIV-protease inhibitors

The effects of HAART on tumour incidence and regression have been most frequently studied in cohorts or groups of patients treated with PI–HAART rather than NNRTI–HAART9,84,103,104,105,106,107,108,109,111,112,114,115,116. This reflects, at least in part, the fact that PI–HAART was in use before the advent of NNRTI–HAART. However, this might also indicate that the unexpected antitumour effects of HAART are related to specific actions of HIV-PIs. This was addressed by directly comparing the impact of PI–HAART and NNRTI–HAART on patients with Kaposi's sarcoma or NHLs. In three recent studies, both regimens were found to be equally effective in reducing the incidence of Kaposi's sarcoma or NHLs14,16,125. Contrasting results, however, have been obtained when looking at tumour regression or relapse. A recent study analysed the response of Kaposi's sarcomas in a series of ten patients treated with PI–HAART or NNRTI–HAART. Both regimens were found to induce partial regression of Kaposi's-sarcoma lesions with a similar frequency126. However, complete remission of Kaposi's sarcoma occurred more frequently in patients treated with PI–HAART than in those treated with NNRTI–HAART126. These patients had very low CD4+ T-cell counts at the beginning of the study, and only small and partial CD4+ T-cell recovery after therapy. So, in patients with low immune activity, HIV-PIs were more efficient at inducing complete response than NNRTI–HAART.

Two other small studies reported the effects of switching patients from PI–HAART to NNRTI–HAART after resolution of Kaposi's sarcoma. Although one study reported that switching these drugs had no effect on relapse, the other study reported that it was consistently associated with tumour relapse, even though both regimens were equally effective against HIV infection127,128. Although larger retrospective and prospective studies are required to draw a firm conclusion, these data indicate that the antitumour effects of HAART might, at least in part, be related to specific actions of HIV-PIs.

Many studies have therefore been aimed at identifying non-virological actions of the most widely used HIV-PIs, including ritonavir, saquinavir, indinavir and nelfinavir. They have found that HIV-PIs directly affect several pathways involved in tumour-cell proliferation and survival, angiogenesis, invasion, inflammation, and antitumour immunity in HIV-free models (Tables 2,3). For example, ritonavir and saquinavir inhibited the proliferation of tumour cell lines of lymphoblastoid origin, including lymphoma cells and myeloid leukaemia cells, fibrosarcoma and mastocytoma cells, as well as immortalized Kaposi's-sarcoma cell lines129,130. This effect was associated with the induction of apoptosis in tumour cells, but no effect on proliferation or survival was observed with non-tumour cells, including non-transformed immortalized fibroblasts or primary macrophages129,130. Furthermore, saquinavir induced apoptosis in prostate cancer cells, glioblastoma cells and lymphocytic leukaemia cells, and sensitized tumour cells to cell death by ionizing radiation21 (Table 2).

Table 2 Direct effects of HIV-PIs on tumours
Table 3 Indirect effects of HIV-PIs on tumours

HIV-PIs also inhibit angiogenesis and tumour growth by blocking cell invasion (Table 2). In particular, studies showed that indinavir and saquinavir inhibited the development of Kaposi's-sarcoma-like angioproliferative lesions induced in nude mice following injection of primary Kaposi's sarcoma cells derived from human lesions22. In treated mice, these drugs promoted the formation of a large central necrotic area and a marked reduction of neoformed vessels, oedema and spindle-cell infiltration22. These effects of HIV-PIs were found to be due to inhibition of basic fibroblast growth factor (bFGF)- and vascular endothelial growth factor (VEGF)-mediated angiogenesis; both proteins are produced by Kaposi's-sarcoma cells22,50,58,59,131,132. Indinavir and saquinavir have also been shown to inhibit the formation of angiogenic lesions directly induced by angiogenic factors in nude mice, as well as bFGF or VEGF-induced angiogenesis in the chicken choriallantoic membrane assay22. However, neither drug had any effect on bFGF-promoted proliferation, basal growth or survival of macrovascular and microvascular endothelial cells, smooth-muscle cells or Kaposi's-sarcoma cells (Table 2). But both drugs completely blocked bFGF-induced invasion of a reconstituted basement membrane (Matrigel) by all these cell types22.

As tumour angiogenesis and invasion are general pathways involved in tumour progression, HIV-PIs might successfully be used to inhibit the growth of tumours of various origins and histotypes. In agreement with this hypothesis, indinavir and saquinavir also blocked tumour formation induced in nude mice by injection of EA-hy 926 cells — a hybrid between human endothelial cells and a lung adenocarcinoma cell line that is used as an angiogenic-tumour model22. Inhibition of these tumours was associated with inhibition of cell invasion, but not cell proliferation or cell survival22. Ongoing studies indicate that indinavir and saquinavir are also effective at inhibiting the growth of various human tumour xenografts, including lung, breast, hepatocarcinoma and colon adenocarcinoma, and human tumours of haematopoietic cell origin (myelomonocytic or T-cell acute leukaemia cells). These effects have also been shown to be mediated by a blockade of tumour angiogenesis and tumour-cell invasion, in the absence of effects on cell proliferation and viability (B.E., unpublished observations).

HIV-PIs exert other activities that might affect tumour-associated inflammation and tumour immunity in HIV-free models. For example, ritonavir and saquinavir inhibit the production and/or release of inflammatory cytokines and chemokines including TNFα, IL-6, and IL-8, by both peripheral-blood mononuclear cells and endothelial cells130,133 (Table 3). This effect of HIV-PIs on inflammatory cytokines has been confirmed in treated patients, as PI–HAART has also been shown to inhibit TNFα, IL-2 and IFNγ production by peripheral-blood mononuclear cells from uninfected individuals who were treated with HIV-PIs for prophylactic intervention without acquiring HIV infection134. Similarly, ritonavir inhibits the expression by endothelial cells of adhesion molecules, including VCAM1, ICAM1, and selectin E, which are known to mediate leukocyte recruitment at sites of inflammation130 (Table 3). These cytokines, chemokines and adhesion molecules are crucial in the development of Kaposi's sarcoma, as they mediate local inflammatory and immune responses to Kaposi's-sarcoma cells and to other KSHV-infected cells. Furthermore, they regulate survival, growth, invasion and eradication of most tumours. In fact, they lead to local stroma activation, basement-membrane and/or extracellular-matrix perturbation, and angiogenesis, and regulate local tumour immunity135.

In this context, HIV-PIs directly modulate antigen processing, T-cell survival and proliferative responses20,136,137,138, and they might even affect T-CELL PRIMING, as they can inhibit dendritic-cell maturation and function139 (Table 3). In an in vivo model of infection by lymphocytic choriomeningitis virus, ritonavir has been shown to modify responses by CYTOTOXIC T LYMPHOCYTES (CTLs) to the virus by inhibiting the processing of viral antigens, although another study reported no alterations in the processing of commonly recognized HIV epitopes20,140. These data indicate that ritonavir might selectively modify the processing of certain antigens. So, ritonavir (and other HIV-PIs capable of affecting antigen processing) could have important effects on the generation of the tumour epitope repertoire, and therefore on the eradication of tumour cells by the immune system (Table 3).

Together, these data indicate that HIV-PIs directly block tumour development and progression as well as tumour metastasis by inhibiting tumour-cell survival, proliferation and invasion, and tumour angiogenesis, and by modulating immunity (Fig. 1).

Figure 1: Steps in tumour progression and metastasis affected by HIV-protease inhibitors.
figure 1

Tumour pathways that underlie the various steps of cancer development can be disrupted by human immunodeficiency virus protease inhibitors (HIV-PIs). These steps usually lead to progression of in situ carcinoma (a) to invasive cancer (b) and to metastasis formation and dissemination (cf). Tumour outgrowth (a,b) is dependent on tumour neoangiogenesis and its net rate is determined by the balance between tumour-cell proliferation versus apoptosis, invasive behaviour and the ability of tumour cells to evade the immune response. At concentrations similar or above therapeutic peak levels, HIV-PIs promote apoptosis and inhibit proliferation of tumour cells with little or no effects on survival and proliferation of normal cells, whereas at therapeutic steady-state concentrations they inhibit tumour angiogenesis and tumour-cell invasion. Furthermore, HIV-PIs have anti-inflammatory effects (b). As metastatic cell clones emerge, tumour cells loosen their contact with surrounding cells and the extracellular matrix (ECM). This leads to invasion of blood or lymphatic vessels and to extravasation of tumour cells at distant sites (ce). These steps require the degradation of basement membranes and, at the same time, inhibition of apoptosis following loss of cell anchorage (anoikis) — processes that are also inhibited by HIV-PIs. Finally, colonization of ectopic tissue by tumour cells (f) is required for establishment of metastases, and this process is similarly affected by HIV-PIs. During tissue invasion and establishment of metastases (b,f), activated endothelial cells, stromal cells and immune cells cooperate in basement membrane and ECM degradation, modify the ECM composition, release ECM-bound growth and angiogenic factors, and produce cytokines and chemokines that stimulate tumour-cell growth and migration, and recruit all these cell types at the invasive front. These processes are all affected by HIV-PIs through their ability to inhibit cytokine and chemokine production, cell activation, and basal membrane and ECM degradation and remodelling.

Mechanisms of action

HIV-PIs differentially affect several tumour pathways, depending on the drug concentration used. For example, at concentrations similar to the lowest levels found in plasma of treated patients (that is, 0.1–1.0 μM), saquinavir and indinavir effectively block invasion of a basement membrane (Matrigel) by endothelial and Kaposi's-sarcoma cells, as well as by lung, breast, colon adenocarcinoma, and myelo-monocytic and T-cell acute leukaemia cells, with no effects on cell survival or proliferation (Ref. 22 and B.E., unpublished observations). At drug concentrations above the therapeutic peak level (50–100 μM), however, saquinavir causes high levels of apoptosis in tumour cells of several tumour types, including prostate cancer, lymphoblastoid leukaemia and glioblastoma21. Both indinavir and saquinavir, used at drug concentrations that are too low to achieve consistent HIV inhibition (10−2–10−1 μM), increase the survival of T cells isolated from HIV-infected patients, and restore antigen-specific proliferative responses to these cells137. Similarly, ritonavir decreases the rate of apoptotic cell death in T cells and in CD34+ haematopoietic-cell progenitors from HIV-non-infected individuals when used at very low drug concentrations (5 × 10−3 to 10−2 μM), an effect that is most likely mediated by a decrease in the expression of both caspases and FAS ligand133,138,141. By contrast, at higher concentrations (5–10 μM) indinavir and ritonavir block T cells from both HIV-infected and non-infected individuals in the G0/G1 or G1/S phase of the cell cycle, and induce necrotic or apoptotic death of T cells and endothelial cells129,136,138,141,142.

Proteasome inhibition. How could protease inhibitors mediate all these antitumour effects and affect all these cell pathways (Fig. 2)? These drugs have been shown to function by inhibiting the activity of the proteasome. This effect occurs at a relatively wide range of drug concentrations (5–100 μM), which are similar to, or above, the pharmacokinetic peak level present in the sera of treated patients20,21,129,130,143,144,145. Within this range, ritonavir, indinavir and saquinavir have been shown not only to inhibit, to various degrees, tryptic, chymotryptic and peptidyl-glutamyl activity of isolated 26S and/or 20S proteasome complexes, but also the function of the 26S proteasome in cultured cells, although for indinavir these effects require combination with NRTIs20,21,129,130,143,144,145. The proteasome controls several cell pathways, including protein turnover, clearance of misfolded proteins, apoptosis, degradation of tumour-suppressor gene products, the function of cyclin-dependent kinase (CDK) inhibitors, and the proteolytic maturation and activation of the transcription factor NF-κB146,147,148. The ability of HIV-PIs to block these activities can inhibit tumour-cell survival and proliferation, as well as tumour-associated inflammation and endothelial-cell activation. By inhibiting the proteasome, HIV-PIs can also sensitize tumour cells to ionizing radiation146,147,148,149. Studies have shown that HIV-PIs act as reversible proteasome inhibitors129,150, much like some potent proteasome inhibitors already being tested in in vitro preclinical studies and in clinical trials as anticancer agents, such as bortezomib or TMC-95 (Refs 147,151).

Figure 2: Molecular targets of antiviral agents.
figure 2

Human immunodeficiency virus protease inhibitors (HIV-PIs) block several signalling pathways that regulate processes such as cell proliferation, survival, activation and invasion. Ritonavir inhibits the expression of FAS ligand (FAS-L) and tumour-necrosis factor-α (TNFα). Signalling by these molecules normally occurs through their receptors (FAS-R, which is also downregulated by ritonavir, and TNF-R, respectively) and downstream molecules, such as FADD and TRADD, to activate caspases (caspases-3, -6 and -7) that induce cell death. TRAF2, however, is also activated by TNFα, which promotes cell survival and proliferation through NF-κB. Inhibition of FAS-R and FAS-L would promote survival of activated leukocytes, which might prevent tumour immune evasion. By contrast, inhibition of TNFα production by activated lymphocytes130 might inhibit NF-κB activation through TRAF2, inhibiting tumour-cell survival. Both ritonavir and saquinavir inhibit or modulate 26S proteasome activity, preventing IκB degradation and thereby blocking NF-κB activation. Ritonavir has also been shown to block NF-κB activation in response to inflammatory cytokines (TNFα signalling) and viral products, including the HIV-1 Tat protein or the gene product of Kaposi's-sarcoma-associated herpesvirus ORF74, which are known to contribute to Kaposi's sarcoma130. Through these actions, these HIV-PIs inhibit tumour-cell survival and proliferation. The ability of these drugs to alter 26S proteasome activity might also modify antigen processing, inhibiting the formation of certain peptide epitopes for presentation to cytotoxic T lymphocytes (CTLs)20,143. These epitopes are normally transported through the endoplasmic reticulum (ER) and presented to CTLs by MHC-class-I (MHC-I) molecules. Alterations in this process could prevent tumour-cell immune evasion by altering the tumour-epitope immunodominance hierarchy. Nelfinavir inhibits apoptosis by preventing loss of mitochondrial-membrane potential and by stabilizing membrane permeability. This prevents the release of cytochrome c and activation other pro-apoptotic molecules, such as apoptotic protease activating factor 1 (APAF1), BAX, BAK, BAD and caspase-9. At low concentrations, saquinavir and indinavir block matrix metalloproteinase 2 (MMP2) proteolytic activation, which requires the binding of MMP2 to integrin22. By contrast, these drugs do not affect the MMP2 pre-activation proteolytic step, which is catalysed by a membrane-associated MMP — membrane type 1 MMP (MT1-MMP). Saquinavir can also inhibit the production and/or release of MMP zymogen (pro-MMP). These effects of indinavir and saquinavir can block angiogenesis and tumour-cell invasion, and might also affect cell survival and inflammation.

For example, saquinavir reversibly inhibits the degradation of ubiquitylated proteins and, through this activity, reversibly sensitizes prostatic tumour cells to ionizing radiation21. Ritonavir, in turn, modulates protein turnover by selectively and reversibly inhibiting the degradation of some, but not all, proteasome substrates, particularly non-ubiquitylated proteins, such as the CDK inhibitor WAF1 (Refs 129,143) (Fig. 2). These actions of ritonavir are due to its capability of selectively binding the MB-1 and LMP7 proteasome subunits143. As LMP7 replaces MB-1 after the generation of the IMMUNOPROTEASOME during inflammatory processes (an effect that is mediated by IFNγ)152, these findings could also explain the capacity of ritonavir to modulate antigen processing and to suppress CTL-dependent inflammatory responses in mice infected by the lymphocytic choriomeningitis virus20. Furthermore, as immunoproteasomes are particularly efficient at generating CTL-epitope peptides, and are constitutively present in PROFESSIONAL ANTIGEN-PRESENTING CELLS, it is possible that HIV-PIs modify the epitope repertoire of tumour antigens. This would have important implications for tumour immunity and tumour control and/or eradication by the immune system (Fig. 2). Although the drug concentrations required to elicit these effects on the proteasome are often well above the pharmacokinetic peak concentration present in treated patients, proteasome modulation at therapeutic drug concentrations could be effective when HIV-PIs are used in combination with NRTIs, such as in HAART regimens. In fact, indinavir has been shown inhibit the 26S proteasome when combined with the NRTIs zidovudine and lamivudine145. Furthermore, inhibition of the proteasome by HIV-PIs has been implicated in the reduced degradation of apolipoprotein B, which, in turn, is associated with HAART-induced hyperlipidaemia153. All these data indicate that HIV-PIs could affect proteasome activity in treated patients.

MMP inhibition. A different mechanism of action, however, is observed at drug concentrations that are too low to affect the cell proteasome, but are similar to the lowest peak level present in the sera from treated patients (0.1–1.0 μM)22. At these lower concentrations, HIV-PIs, including indinavir and saquinavir, inhibit angiogenesis and cell invasion through their effects on the activity of MMPs, particularly MMP2 (Ref. 22). MMPs, or the enzymes and molecules known to regulate their production and/or activity, do not share any sequence homology or structural similarity with the HIV protease22. Furthermore, HIV protease and MMPs do not belong to the same functional class of proteases22. Accordingly, indinavir and saquinavir do not affect the catalytic activity of MMPs in a direct way22. Rather, they inhibit the proteolytic activation of MMP2 with no effects on its production or release22.

MMP2 activation is initiated by proteolytic enzymes, such as membrane type 1 MMP (MT1-MMP), which cleave latent MMP2 to produce the pre-active form of the enzyme (pre-MMP2). In turn, pre-MMP2 is fully activated after autoproteolytic cleavage that is mediated by the αvβ3-integrin22. The exact mechanism for the inhibition of MMP2 activation by HIV-PIs (and whether integrins are direct targets of HIV-PIs) has not yet been determined. However, in vitro studies indicate that activation of purified MMP2 is not affected by HIV-PIs, indicating a role for MT1-MMP, αvβ3-integrin and/or other molecules involved in MMP production or activation in this effect of HIV-PI (B.E., unpublished observations). Furthermore, HIV-PIs affect the production and/or release, but not the activation, of MMP3 (Ref. 154). As MMP production is induced after integrin activation155,156, it is possible that integrins or molecules involved in integrin signalling could be targeted by HIV-PIs.

This effect of HIV-PIs on MMPs, cell invasion and angiogenesis is likely to inhibit tumour growth and metastasis (Figs 1,2), as well as lead to the regression of early tumours, with little effect on already established or advanced tumours. In fact, neoformed and yet non-stabilized vessels, which are the targets of anti-angiogenic agents, are prominent in early but not in advanced tumours. Indeed, PI–HAART promotes regression of Kaposi's sarcoma and prolongs disease-free time in patients with early disease, whereas such responses are only achieved in patients with massive, advanced disease after debulking chemotherapy (Ref. 157 and U. Tirelli, personal communication).

Furthermore, MMPs are involved in several crucial immune and immunomodulatory functions, and in cancer-mediated immune suppression. Evidence indicates, in fact, that MMPs participate in antigen processing158. In addition, recent studies have shown that the direct injection of recombinant MMPs in mice can induce dendritic-cell maturation and trafficking159. Moreover, MMPs, including MMP2, act as potent modulators of local inflammation by activating or degrading inflammatory cytokines and chemokines present on the cell membrane, such as TNFα, IL-1β, monocyte chemoattractant protein 3 and IL-8 (Refs 160164). Importantly, MMPs activate transforming growth factor-β, which, in turn, inhibits T-cell responses against tumours165,166. MMPs can also cleave IL-2 receptor-α167, which is required for T-cell proliferation following antigen stimulation. These effects of MMPs are important determinants of tumour immune evasion, but might also explain the strong stimulatory effect of low HIV-PI concentrations on T-cell proliferation and survival. Furthermore, as MMPs are required for leukocyte transmigration and tissue infiltration by inflammatory cells, the capability of ritonavir to inhibit CTL-dependent inflammatory responses could be mediated not only following the modulation of CTL-epitope processing by the proteasome, but also through the inhibition of MMP activation or function in transmigrating lymphocytes140. Together, these actions on MMPs could explain the ability of PI–HAART to inhibit T-cell activation, inflammatory cytokine production, and inflammation, as well as to modulate dendritic-cell maturation, and might contribute to their antitumour effects in patients.

As HIV-PIs are cleared with very fast kinetics in patients, plasma concentrations of these drugs are only transiently sufficient to affect proteasome function, but remain for long periods of time above the concentration required to block MMPs, cell invasion and angiogenesis. So, blockage of cell invasion and angiogenesis through MMP inhibition is likely to be the most prominent mechanism underlying the antitumour effects of HIV-PIs in patients22. Studies have indicated that PI–HAART inhibits MMPs in humans, as mutations in genes encoding MMP2 or MT1-MMP lead to bone disorders such as OSTEOPAENIA, increased bone resorption and OSTEOPOROSIS168 — effects that are also observed in patients who are treated with PI–HAART169,170.

Future directions

Based on what we have learned about the mechanisms of HIV-PIs, might these drugs be exploited for the therapy of patients with non-HIV-associated cancers? The actions of HIV-PIs on cell survival, apoptosis, cell invasion, angiogenesis, inflammation and immune responses indicate that these drugs could have widespread use in the treatment of all cancer types. In particular, HIV-PIs should be exploited, alone or in combination with conventional cytotoxic therapy, for the treatment of tumours that have been shown to be sensitive to anti-angiogenic therapy, such as colon and renal cancer, and multiple myeloma171, or to prevent the invasion of pre-malignant diseases, such as in situ carcinomas of the oral cavity and cervix. In addition, HIV-PIs might have a favourable therapeutic index in the treatment of tumours that are sensitive to the anti-inflammatory cyclooxygenase (COX) inhibitors, such as gastrointestinal, breast and lung cancer172,173. In fact, COX2 is induced by inflammatory cytokines that, in turn, are inhibited by HIV-PIs173. Because of their anti-angiogenic and pro-apoptotic actions, HIV-PIs might be useful in preventing tumour recurrence or metastasis after conventional radiation or cytotoxic therapy. This is also indicated by the increased survival of patients with AIDS-associated tumours treated with HAART combined with chemotherapy174.

It is, therefore, important to determine the specific interactions of HIV-PIs with conventional cytotoxic drugs. In recent studies, PI–HAART has not shown significant interactions with doxorubicin or paclitaxel175,176, but it has been reported to increase the haematological and neurological toxicity of chemotherapy177,178. This, however, could be due to NRTIs, which are known to exert such toxic effects177. In this context, HIV-PIs have been shown to inhibit apoptosis of haematopoietic precursors and polymorphonuclear leukocytes (PMNL) and to stimulate haematopoiesis and PMNL function141,179,180,181. So, when used in uninfected individuals, HIV-PIs might decrease, rather than increase, the haemotological side effects of cytotoxic drugs. In addition, as ritonavir inhibits the functional activity of multidrug-resistance-related protein 1, it might be exploited to overcome resistance to anticancer drugs182.

The relatively low toxicity of these drugs and the large body of data on their pharmacokinetics, tissue distribution and drug interactions would allow for the rapid clinical evaluation of HIV-PIs alone or combined with other molecules in patients with cancer. To this end, a Phase II clinical trial for the treatment of classical Kaposi's sarcoma in HIV-uninfected individuals with indinavir, and other clinical studies for the treatment of other tumour types with HIV-PIs, are underway in Italy to asses the efficacy of HIV-PIs, alone or in combination with classic cytotoxic drugs, in HIV-positive and HIV-negative cancer patients (Table 4). Surrogate markers of angiogenesis, tumour invasion, immune activation, and TH1 and TH2 responses could be used to validate the actions of these drugs in patients (Table 4). It is also of great importance to better dissect the anti-angiogenic and antitumour actions of the new NNRTI-based regimens, compared with PI–HAART, to determine if these will be as effective in the management of HIV-infected patients with tumours or who are at risk of tumours. Further basic and preclinical studies to compare the antitumour activity of NNRTIs versus HIV-PIs in non-HIV-associated models are needed, as well as retrospective and prospective studies aimed at dissecting the non-antiretroviral actions of NNRTIs in HIV-infected patients (Table 4). Finally, a complete exploitation of HIV-PIs as antitumour drugs requires further definition of their exact mechanism of action. The integration of functional genomics and proteomics techniques in the study of the effect of HIV-PIs on tumour behaviour; molecular modelling and docking approaches; and protein–ligand interaction studies are required to identify the exact targets of HIV-PIs, and for developing new HIV-PI derivatives and analogues with higher therapeutic indexes.

Table 4 Clinical studies/trials underway to evaluate the effects of HIV-PIs on cancer

Recent studies have indicated that the proteasome might regulate angiogenesis and cell invasion by modulating MMP expression and/or activation as well as angiogenic-factor receptors183,184,185,186. So, there might be crosstalk between the two key pathways that are targeted by HIV-PIs. Studying the effects of HIV-PIs on tumour cells could shed light on such a connection, leading to new avenues in drug discovery based on the rational design of a new class of antitumour drugs based on HIV-PIs.