Skip to main content

MINI REVIEW article

Front. Immunol., 21 March 2017
Sec. NK and Innate Lymphoid Cell Biology
This article is part of the Research Topic NK Cells in Human Diseases: Friends or Foes? View all 12 articles

Role of Natural Killer Cells in HIV-Associated Malignancies

  • 1Programa de Oncovirologia, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
  • 2Department of Tropical Medicine, Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
  • 3Blood Systems Research Institute, San Francisco, CA, USA
  • 4University of California, San Francisco, CA, USA

Now in its fourth decade, the burden of HIV disease still persists, despite significant milestone achievements in HIV prevention, diagnosis, treatment, care, and support. Even with long-term use of currently available antiretroviral therapies (ARTs), eradication of HIV remains elusive and now poses a unique set of challenges for the HIV-infected individual. The occurrence of HIV-associated non-AIDS-related comorbidities outside the scope of AIDS-defining illnesses, in particular non-AIDS-defining cancers, is much greater than the age-matched uninfected population. The underlying mechanism is now recognized in part to be related to the immune dysregulated and inflammatory status characteristic of HIV infection that persists despite ART. Natural killer (NK) cells are multifunctional effector immune cells that play a critical role in shaping the innate immune responses to viral infections and cancer. NK cells can modulate the adaptive immune response via their role in dendritic cell (DC) maturation, removal of immature tolerogenic DCs, and their ability to produce immunoregulatory cytokines. NK cells are therefore poised as attractive therapeutic targets that can be harnessed to control or clear both HIV and HIV-associated malignancies. To date, features of the tumor microenvironment and the evolution of NK-cell function among individuals with HIV-related malignancies remain unclear and may be distinct from malignancies observed in uninfected persons. This review intends to uncouple anti-HIV and antitumor NK-cell features that can be manipulated to halt the evolution of HIV disease and HIV-associated malignancies and serve as potential preventative and curative immunotherapeutic options.

Introduction

Since the introduction of antiretroviral therapy (ART), life expectancy of people living with HIV (PLHIV) has notably improved, and the gap between the uninfected population ranging from 60 to 90%, of normal life expectancy is narrowing in regions of the world among those with access to ART (1). Before the availability of ART, immune suppression-related complications represented the predominant cause of mortality among HIV-infected individuals. Incidence rates of non-Hodgkin lymphoma (NHL) and Kaposi sarcoma (KS) were more than 100 times higher in the pre-ART era and were classified, together with cervical cancer, as AIDS-defining cancers (ADCs) (2). Overtime, as ART became the standard of care, prolonged use has lead to a remarkable improvement in immune status, dramatically reducing ADC rates (ratio of ART to no-ART) by 0.61 per year (3).

In the United States, despite the sharp decline in the incidence of ADCs, increased risk of developing specific types of NHL such as Burkitt lymphoma and classical Hodgkin lymphoma (HL) has evolved (4, 5). A meta-analysis of standardized incidence ratios from 18 studies showed that infection-related cancers such as anal, liver and HL, as well as smoking-related cancers such as lung, kidney, and oral cancers, had an increased incidence among the HIV population despite being on ART, with lymphomas being the most frequent type of cancer observed (6). A separate comprehensive epidemiological study showed that the PLHIV also had significantly higher rates of these now termed “non-AIDS defining cancers” (NADCs) when compared to matched HIV-negative individuals, suggesting the likelihood of other etiologies besides an increase in life expectancy as predisposing the HIV population to develop these types of cancers more frequently (7). In fact, NADCs have now become one of the leading causes of mortality among HIV-infected individuals (8, 9). Surprisingly, however, the incidences of breast and prostate cancers have significantly declined in HIV-infected persons, suggesting that HIV infection, ART, or other viral–host interactions have differential impacts on cancer risk in this population (10, 11). The increased incidence of NADC despite viral suppression and CD4 T-cell recovery in the era of ART raises important key mechanistic questions in the oncogenesis of NADC. The direct oncogenic effect of HIV, HIV-induced immunodeficiency, and chronic inflammation, as well as ART toxicities are some of the plausible mechanisms that are being investigated (12). Complete immune recovery after prolonged ART is variable and underscores that harnessing specific components of the host immune response may play a vital role in preventing NADC.

Chronic immune activation and immune senescence contribute to immune dysfunction in chronic HIV infection and partially persist even after CD4 T-cell count recovery and viral suppression by ART (13). Such processes lead to immune exhaustion/senescence, thereby facilitating reactivation of other latent viral infections, such as Epstein–Barr virus (EBV). Besides HIV, all ADCs and the majority of NADCs appear to be associated with several chronic viral infections (12), justifying a new way to categorize HIV malignancies into infection-related and non-infectious-related cancers. KS is intimately associated with HHV-8 infection (14), and cervical cancer is almost always caused by HPV infection (15). B-cell lymphoproliferative disorders are frequently associated with EBV infection, and such association is even more common in HIV-infected individuals, ranging from 60 to 100% (5, 16). Viral co-infections are present in NADC. The incidence of hepatocellular carcinoma has progressively increased among HIV-infected persons in the last decade (17). Merkel cell carcinoma, recently reported to be associated with Merkel cell virus, has a 20-fold increased risk among HIV-infected individuals (18). The potential direct effects of HIV in modulating oncogenes are under investigation, but how HIV impacts the oncogenic potential of other chronic viral infections is unclear (1921). Persistent immune alterations may play a critical role in the oncogenic process in this population and deserve special attention, particularly in the context of co-infections.

Natural Killer (NK) Cells: A Critical Immune Player in Antitumor and Anti-HIV Immunity

Since the discovery of NK cells 40 years ago, a plethora of research has uncovered their phenotypic and functional capacity against virally infected and tumor cells (2225). NK cells are CD3− multifunctional effector lymphocytes defined based on levels of CD56 and CD16 expression (26), the majority (>90% of NK cells) in the peripheral blood being CD56dim and predominantly cytotoxic upon activation, thereby releasing pro-apoptotic cytoplasmic granules composed of granzymes and perforins. CD56dim NK cells can also induce cytolysis via induction of Fas/FasL-dependent or TRAIL-dependent apoptotic signals. In addition, a minority of NK cells express the FcγRIIIA receptor (CD16) that binds to the constant (Fc) domain of IgG antibodies that can bind to viral antigens expressed on the surface of infected cells. This antibody conjugation of NK-cell and antibody-coated target cell, strongly mediating NK-cell activation, is known as antibody-dependent cell-mediated cytotoxicity (ADCC) (27). A distinct subset of CD56bright cytokine-producing NK cells with a limited cytotoxic capacity is more abundantly present in lymph nodes (28). By producing IFN-γ, TNF-α, IL-10, and chemokines, this NK subset predominantly modulates other subsets of lymphocytes, thereby regulating dendritic cell maturation, differentiation of helper T cells, and B- and T-cell-specific immune responses (29, 30).

To understand the NK-cell effector functions, it is paramount to take into consideration the balance between activating and inhibitory signals (31) that drive NK-cell cytotoxicity. NK-cell activation relies on stimulatory signals capable of overcoming the steady inhibitory state that is maintained by signaling through inhibitory receptors. Self-recognition of MHC-I proteins through C-type lectin receptor NKG2A and inhibitory killer cell immunoglobulin-like receptors (KIRs) represent the physiological interaction between NK and target cells. The absence of recognition of “self” by inhibitory receptors characterizes the “missing-self” phenomenon and lowers the activating threshold. NK cells become more susceptible to activation, especially if activating molecules are expressed in infected or transformed target cells and recognized by activating receptors, characterizing the altered-self phenomenon. Activating C-type lectin receptor NKG2D recognizes the altered self-state of infected or transformed cells and triggers NK-cell cytolytic activity. Other surface molecules, such as natural cytotoxic receptors Nkp30, Nkp44, and Nkp46, and activating KIRs also contribute to NK-cell activation process and are critical to determine whether NK cells will be activated to target infected or transformed cells (27, 31).

Both HIV infection and oncogenesis lead to a downregulation of surface MHC-I expression as a way to avoid T-cell recognition but in turn renders target cells more susceptible to NK-cell-mediated cytolysis. However, HIV has developed immune evasion mechanisms via the viral protein Nef, thereby leading to preferential downregulation of HLA-A and -B, and preserving expression of HLA-C and -E (32). Therefore, HIV prevents NK activation as well as CTL recognition of infected cells. Besides interfering with self-recognition, HIV infection and cancer can induce expression of stress signaling molecules, in particular MHC class I polypeptide-related sequence A/B (MICA/MICB). More importantly, HIV leads to persistent activation and consequently T cell and NK-cell immune exhaustion. Despite viral suppression and normal CD4 T-cell counts in the majority of HIV-infected persons on ART, NK-cell phenotype and functionality are not fully restored, suggesting that these individuals may be more susceptible to long-term comorbidities associated with immune dysfunction, such as HIV-related malignancies (33).

The Interplay between the Tumor Microenvironment and NK-Cell Immunity

The process by which the immune system can promote or suppress tumor growth and development is based on animal models and data from cancer patients and has evolved to define the concept of cancer immunoediting (34). Tumor immunoediting is comprised of three phases: elimination, equilibrium, and escape. The elimination phase is when immune cells target cancer cells that succeeded in overcoming intrinsic tumor suppressor mechanisms. If tumor elimination is only partially achieved, a state of equilibrium between malignant cells and the immune system ensues. Tumor cells can become dormant or accumulate mutations, while the immune system continues to exert selective pressure, thereby controlling tumor progress temporarily or eventually eliminating the cancer cells. If elimination does not occur, tumor cell variants resistant to the existent immune response eventually give rise to tumor progression, thereby initiating the escape phase and characterizing failure of tumor immune control. The contribution of NK cells in cancer immunoediting and clinical outcomes is now being appreciated (35).

Natural killer cells have proved to be critical for the eradication and inhibition of metastasis of cancer cells in vivo (36). Perforin protein (pfp) and/or IFN-γ knockout (KO) mice predominately develop B-cell lymphomas, especially after 1 year of age (older animals) with a combination of pfp and IFN-γ KO inducing an early onset of lymphoma, suggesting a synergistic immunosurveillance effect (37). Late age development of B-cell lymphoma and lung adenocarcinoma were also observed in TRAIL KO mice (38) and pfp KO mice (37), respectively. These findings support a role for NK-cell immunosurveillance of B-cell lymphomas as well as epithelial malignancies, through a combination of NK-cell-mediated cytotoxic activity, IFN-γ secretion, and TRAIL killing pathways. In humans, NK cells are particularly relevant in the prevention of tumor development. An 11-year follow-up of the general population for cancer incidence showed an association between reduced NK-cell cytotoxicity and increased risk of cancer (39). It has been postulated that NK cells are critical to the prevention of cancer development (elimination and equilibrium); however, once the tumor microenvironment is established (escape), suppressor factors such as TGF-β and IL-10 are induced and negative inhibitory receptors, such as the T-cell immunoglobulin-and-mucin-domain-containing molecule-3 receptor (TIM-3) on NK cells, that maintain an NK-cell anergic state (40) are upregulated. The induced aberrant expression of HLA-G (membrane-bound and soluble) and increased shedding of MICA (sMICA) seen in tumor cells (41, 42) can further suppress NK-cell antitumor immune responses. HLA-G interaction with ILT2 and CD94/NKG2A results in the inhibition of NK-cell cytotoxicity, IFN-γ secretion, and chemotaxis (43), while sMICA–NKG2D binding impairs NK-cell tumor-specific cytotoxicity, NKG2D expression, and homeostatic maintenance (42).

As a result of these immune deregulated events, NK-cell-associated suppressor factors are currently being considered as immunotherapeutic targets. TIM-3, for instance, is an immunoregulatory checkpoint expressed by most lymphocyte subtypes with critical and complex implications in cytotoxic NK cells (44, 45). Increased TIM-3 expression on NK cells has been shown as a marker of poorer prognosis in lung adenocarcinoma and other types of cancer and correlates with reduced NK-cell cytotoxicity. Blockade of TIM-3 is capable of restoring IFN-γ secretion and cytotoxicity of NK cells in lung cancer (46). Recent studies by Fowke et al. have shown that low expression of various inhibitory molecules on NK cells were associated with HIV viral control (47). Despite the complexity of the immune suppressive strategy of the tumor microenvironment, targeting these inhibitory checkpoint receptors shows potential to restore NK-cell functionality in the control or clearance of solid tumors (48). Currently, several trials are underway assessing the impact of Tim-3 blockade in cancer patients (http://ClinicalTrials.gov: NCT02817633; NCT02608268). However, such therapeutic tools are still in their infancy in the context of HIV-associated ADC or NADCs. Given the success of immunotherapy targeting the inhibitory receptors PD-1 and CTLA-4 against several malignancies (49), evaluation of the impact of NK-cell function following immune checkpoint blockade may have relevance in the setting of HIV and may serve a dual purpose in both HIV eradication and tumor clearance. Combining immunotherapy and NK-cell-based therapies is another potential targeted strategy and warrants further investigation in individuals with HIV-associated malignancies (5053).

NK-Cell Immune Control of HIV Infection during ART

HIV infection induces significant phenotypic changes and negatively impacts NK-cell cytotoxicity (54). Cytotoxic NK cells in aviremic HIV donors have impaired ADCC that is associated with a reduced expression of FcRIIIA, an activated phenotype represented by increased expression of CD38 and HLA-DR. Furthermore, NK cells are rarely found in lymph nodes, an important site of both HIV replication and B-cell transformation (55). Critical activating receptors important in cancer immunosurveillance such as NKp30 and NKp46 are downregulated (56) in HIV infection, and expansion of dysfunctional CD56− NK-cell subsets (57) persists even after cART.

Non-neutralizing anti-HIV-1 antibodies can mediate protection through ADCC in assays of HIV candidate vaccines in non-human primate models of HIV infection. Several studies have suggested that HIV-specific ADCC responses may be contributing to partial control of HIV viremia during acute infection. The early initial interest in the utility of NK-mediated ADCC via HIV-specific antibodies was to enhance the development of an HIV vaccine and novel therapies to suppress HIV replication. Interestingly, in the RV144 HIV vaccine Thai trial, the robust ADCC responses observed were associated with modest protective efficacy (58). It would be intriguing to determine the effects of NK-mediated ADCC in the recently launched HVTN 702 study that builds on the success of the RV144 trial in support of this correlate. In addition, recent studies suggest that cytokine stimulation can enhance direct NK cytotoxicity and NK-mediated ADCC of autologous HIV-infected CD4+ T cells (5961). Therefore, modulating NK activity is a potential strategy to develop novel immunotherapies to prevent and possibly lead to the eradication of HIV.

Influence of NK-Cell Immunity in HIV-Associated Malignancies

It is becoming apparent that NK cells may also contribute to tumorigenesis. The potential impact of such alterations in HIV malignancies is illustrated in Figure 1. Overexpression of activation markers on NK cells and spontaneous degranulation occurring during HIV infection may directly contribute to tumor development (33). Analysis of NK cells from patients with lymphoma demonstrated decreased levels of activating receptors in those with HIV compared to uninfected patients, suggesting that these cells might be less efficient to target cancer cells (62). NK cells can also present proangiogenic activity in the tumor microenvironment in a similar way to decidual NK (dNK) cells early on in pregnancy. In fact, in non-small cell lung cancer, the majority of tumor-infiltrating NK cells have a dNK-cell phenotype: CD56bright, CD9+, perforin low, and high production of vascular endothelial growth factor (VEGF) (63). Hypoxia and TGF-β secreted by tumor cells has a known immunomodulatory impact in the tumor microenvironment and induces VEGF secretion (64). In vitro exposure to TGF-β and hypoxia led to conversion of CD56dim NK cells into dNK-like cells (65). HIV infection leads to increased levels of TGF-β by monocytes (66) and T cells (67), suggesting that TGF-β may play a more prominent role in tumorigenesis during HIV infection.

FIGURE 1
www.frontiersin.org

Figure 1. Compounded effect of HIV infection on natural killer (NK) cell antitumor responses. The tumor microenvironment constrains NK-cell functionality through the expression of tumor-derived transforming growth factor β (TGF-β) (68), shedding of MICA, and HLA-G (69). The limitations of antitumor mechanisms by NK cells are exacerbated in HIV infection. HIV infection reduces the surface expression of activation receptors (aKIRs, NKp30, and NKp46) and CD16 (56) while upregulating the expression of inhibitory receptors (iKIRs). The net result of the influence of HIV on NK-cell receptor expression further impairs NK-cell activation by cancer cell interaction and decreases tumor-directed antibody-dependent cell-mediated cytotoxicity (ADCC) responses (70). HIV infection decreases INF-γ and TNF-α production by NK cells despite HIV viral suppression by cART (71, 72), which will limit dendritic cell (DC) maturation and thus prevent efficient tumor-directed adaptive responses (73). The increased plasma TGF-β and loss of cell-specific degranulation of NK cells seen in HIV infection could lead to tumorigenicity via contributing to increased frequency of vascular endothelial growth factor (VEGF)-producing intratumoral NK cells (63) and occurrence of chronic inflammation, respectively (74). Furthermore, the affect of cART on tumor activity is yet to be explored. Blue lines represent responses that promote tumor growth, while responses that inhibit NK-cell function are indicated in red. Decreases are indicated by dashed lines and increases by bolded lines.

The combination of (1) reduced expression of activating receptors and increased inhibitory receptors (e.g., TIM-3), (2) reduced ADCC, (3) reduced secretion of TNF-α and IFN-γ, and (4) development of pro-cancer features such as persistent activation, spontaneous degranulation, and production of VEGF suggests that NK cells may directly be associated with the increased cancer risk in the setting of ART-treated HIV infection. It is fair to speculate that targeted immunotherapies reversing specific NK cells deficits may be relevant for many HIV-related malignancies.

NK-Cell-Based Immunotherapies Targeting HIV and HIV-Associated Malignancies

With the exception of the Berlin patient (75), and given the continued resurgence of virus in HIV-infected persons in various eradication approaches (7678), it is clear that elimination of all latent HIV reservoirs is going to be critical to successfully achieve ART-free sustained HIV control or remission. Innovative approaches that are extrapolated from these studies and cases have lead to renewed interest in determining ways to bolster the host immune response and/or manipulate HIV target cells to render them refractory to infection. Since HIV and cancers have evolved sophisticated modalities to escape the host immune defense mechanisms, enhancing NK-cell function may serve as a promising tool as part of a multifaceted approach in the elimination of HIV as well as HIV-associated malignancies.

Recently, there has been renewed interest in harnessing HIV-specific ADCC responses as an HIV cure strategy. A monoclonal antibody (mAb) targeting the CD4-binding site on the HIV envelope spike (3BNC117) may have the potential to guide host immune effector cells to accelerate the clearance of HIV-1-infected cells by an FcyR-dependent mechanism (79, 80). In addition, Byrareddy et al. recently reported that a rhesusized mAb against α4β7 mediated sustained control of SIV infection in the absence of ART in non-human primates. This remarkable response was associated with increased cytokine-synthesizing NK cells (81). These studies, and others, highlight the potential of using mAbs through modulation of NK-cell-mediated activity as an exciting therapeutic tool to achieve sustained HIV remission and be beneficial in the context of HIV-associated tumors.

Natural killer-cell-based antitumor immunotherapeutic strategies targeting NK-cell activity have shown some promise in the oncology field (Table 1). Infusion of allogeneic or autologous NK cells has, to some degree, been successful in tumor clearance. Recently, single-chain variable fragment recombinant reagents, such as bispecific and trispecific killer cell engagers, are being considered as novel immunomodulators to enhance NK-cell function, antigen specificity, and in vivo expansion of these infused cells; however, these reagents need to be fully evaluated for clinical use (8291). The efficiency of ADCC-mediated NK-cell responses is dependent on several factors from the mAbs themselves, NK-cell, and target cell status and also to the glycosylation state and the expression of glycosylation-specific ligands of both the NK-cell and target cells (92, 93). These features can be modified to enhance antiviral and immunomodulatory functions and/or the ability of the target cell to trigger or evade immunological recognition.

TABLE 1
www.frontiersin.org

Table 1. Therapeutic strategies utilizing natural killer (NK) cells in cancer immunotherapy that should be evaluated in the eradication of both HIV and HIV-associated malignancies.

Several gene-editing technologies have been explored to genetically reprogram NK cells to optimize their persistence, expansion, migration, and cytotoxic capacity to improve the antitumor efficacy of primary human NK cells in vivo. With many challenges associated with most of these technologies, CRISPR/Cas9 nuclease system offers a new promising tool to gene-edit NK cells to improve their utility as a novel cell-based cancer immunotherapy strategy (142). Finally, latency reversal agents (LRAs) are being explored as a part of a “shock” approach to reverse cellular HIV latency and expose HIV reservoirs to immune-mediated clearance. Garrido et al. recently studied the impact of LRAs on the function of primary NK cells ex vivo and showed a heterogeneous mixed effect of different LRAs on antiviral activity, cytotoxicity, cytokine secretion, phenotype, and viability (143). Therefore, it will be important to comprehensively evaluate the impact of potential HIV curative strategies and elucidate the possible effect on NK cells against HIV-associated cancers.

Conclusion

To achieve ART-free sustained HIV remission, it is likely that a multidisciplinary approach that includes a combination of immunotherapies and novel targeted approaches that will boost both the adaptive and innate immunity will be necessary. From this review, we posit that NK cells remain a very attractive therapeutic target to develop curative interventions for both HIV and HIV-associated malignancies.

Author Contributions

LN and MA-M conceived and designed the review; LN, MA-M, and FL wrote the paper; and TP contributed to editing the review and designed and developed the figure.

Conflict of Interest Statement

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

Funding

MA-M receives support from the National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIH/NIAID) grant number R21AI129636 and LN receives support from NIH/NIAHD grant numbers P30 AI027757 and R21 AI122393.

References

1. Wandeler G, Johnson LF, Egger M. Trends in life expectancy of HIV-positive adults on antiretroviral therapy across the globe: comparisons with general population. Curr Opin HIV AIDS (2016) 11(5):492–500. doi: 10.1097/COH.0000000000000298

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Goedert JJ, Cote TR, Virgo P, Scoppa SM, Kingma DW, Gail MH, et al. Spectrum of AIDS-associated malignant disorders. Lancet (1998) 351(9119):1833–9. doi:10.1016/S0140-6736(97)09028-4

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Chao C, Leyden WA, Xu L, Horberg MA, Klein D, Towner WJ, et al. Exposure to antiretroviral therapy and risk of cancer in HIV-infected persons. AIDS (2012) 26(17):2223–31. doi:10.1097/QAD.0b013e32835935b3

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Biggar RJ, Chaturvedi AK, Goedert JJ, Engels EA; HIV/AIDS Cancer Match Study. AIDS-related cancer and severity of immunosuppression in persons with AIDS. J Natl Cancer Inst (2007) 99(12):962–72. doi:10.1093/jnci/djm010

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Carbone A, Volpi CC, Gualeni AV, Gloghini A. Epstein-Barr virus associated lymphomas in people with HIV. Curr Opin HIV AIDS (2017) 12(1):39–46. doi:10.1097/COH.0000000000000333

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Shiels MS, Cole SR, Kirk GD, Poole C. A meta-analysis of the incidence of non-AIDS cancers in HIV-infected individuals. J Acquir Immune Defic Syndr (2009) 52(5):611–22. doi:10.1097/QAI.0b013e3181b327ca

CrossRef Full Text | Google Scholar

7. Robbins HA, Pfeiffer RM, Shiels MS, Li J, Hall HI, Engels EA. Excess cancers among HIV-infected people in the United States. J Natl Cancer Inst (2015) 107(4):dju503. doi:10.1093/jnci/dju503

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Zucchetto A, Virdone S, Taborelli M, Grande E, Camoni L, Pappagallo M, et al. Non-AIDS-defining cancer mortality: emerging patterns in the late HAART era. J Acquir Immune Defic Syndr (2016) 73(2):190–6. doi:10.1097/QAI.0000000000001033

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Rodger AJ, Lodwick R, Schechter M, Deeks S, Amin J, Gilson R, et al. Mortality in well controlled HIV in the continuous antiretroviral therapy arms of the SMART and ESPRIT trials compared with the general population. AIDS (2013) 27(6):973–9. doi:10.1097/QAD.0b013e32835cae9c

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Engels EA, Biggar RJ, Hall HI, Cross H, Crutchfield A, Finch JL, et al. Cancer risk in people infected with human immunodeficiency virus in the United States. Int J Cancer (2008) 123(1):187–94. doi:10.1002/ijc.23487

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Grulich AE, van Leeuwen MT, Falster MO, Vajdic CM. Incidence of cancers in people with HIV/AIDS compared with immunosuppressed transplant recipients: a meta-analysis. Lancet (2007) 370(9581):59–67. doi:10.1016/S0140-6736(07)61050-2

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Borges AH, Dubrow R, Silverberg MJ. Factors contributing to risk for cancer among HIV-infected individuals, and evidence that earlier combination antiretroviral therapy will alter this risk. Curr Opin HIV AIDS (2014) 9(1):34–40. doi:10.1097/COH.0000000000000025

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Nasi M, De Biasi S, Gibellini L, Bianchini E, Pecorini S, Bacca V, et al. Ageing and inflammation in patients with HIV infection. Clin Exp Immunol (2016) 187(1):44–52. doi:10.1111/cei.12814

CrossRef Full Text | Google Scholar

14. Dittmer DP, Damania B. Kaposi sarcoma-associated herpesvirus: immunobiology, oncogenesis, and therapy. J Clin Invest (2016) 126(9):3165–75. doi:10.1172/JCI84418

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Senapati R, Senapati NN, Dwibedi B. Molecular mechanisms of HPV mediated neoplastic progression. Infect Agent Cancer (2016) 11:59. doi:10.1186/s13027-016-0107-4

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Carbone A, Cesarman E, Spina M, Gloghini A, Schulz TF. HIV-associated lymphomas and gamma-herpesviruses. Blood (2009) 113(6):1213–24. doi:10.1182/blood-2008-09-180315

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Merchante N, Merino E, Lopez-Aldeguer J, Jover F, Delgado-Fernandez M, Galindo MJ, et al. Increasing incidence of hepatocellular carcinoma in HIV-infected patients in Spain. Clin Infect Dis (2013) 56(1):143–50. doi:10.1093/cid/cis777

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Wieland U, Kreuter A. Merkel cell polyomavirus infection and Merkel cell carcinoma in HIV-positive individuals. Curr Opin Oncol (2011) 23(5):488–93. doi:10.1097/CCO.0b013e3283495a5b

CrossRef Full Text | Google Scholar

19. De Paoli P, Carbone A. Microenvironmental abnormalities induced by viral cooperation: impact on lymphomagenesis. Semin Cancer Biol (2015) 34:70–80. doi:10.1016/j.semcancer.2015.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Dolcetti R, Gloghini A, Caruso A, Carbone A. A lymphomagenic role for HIV beyond immune suppression? Blood (2016) 127(11):1403–9. doi:10.1182/blood-2015-11-681411

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Wang L, Li G, Yao ZQ, Moorman JP, Ning S. microRNA regulation of viral immunity, latency, and carcinogenesis of selected tumor viruses and HIV. Rev Med Virol (2015) 25(5):320–41. doi:10.1002/rmv.1850

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Herberman RB, Nunn ME, Holden HT, Lavrin DH. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. II. Characterization of effector cells. Int J Cancer (1975) 16(2):230–9. doi:10.1002/ijc.2910160205

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Herberman RB, Nunn ME, Lavrin DH. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int J Cancer (1975) 16(2):216–29. doi:10.1002/ijc.2910160205

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Kiessling R, Klein E, Pross H, Wigzell H. “Natural” killer cells in the mouse. II. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Characteristics of the killer cell. Eur J Immunol (1975) 5(2):117–21. doi:10.1002/eji.1830050208

CrossRef Full Text | Google Scholar

25. Kiessling R, Klein E, Wigzell H. “Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol (1975) 5(2):112–7. doi:10.1002/eji.1830050208

CrossRef Full Text | Google Scholar

26. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol (2001) 22(11):633–40. doi:10.1016/S1471-4906(01)02060-9

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Wu J, Lanier LL. Natural killer cells and cancer. Adv Cancer Res (2003) 90:127–56. doi:10.1016/S0065-230X(03)90004-2

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, et al. The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic. J Immunol (2004) 172(3):1455–62. doi:10.4049/jimmunol.172.3.1455

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A, et al. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1 priming. Nat Immunol (2004) 5(12):1260–5. doi:10.1038/ni1138

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Mocikat R, Braumuller H, Gumy A, Egeter O, Ziegler H, Reusch U, et al. Natural killer cells activated by MHC class I(low) targets prime dendritic cells to induce protective CD8 T cell responses. Immunity (2003) 19(4):561–9. doi:10.1016/S1074-7613(03)00264-4

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Cerwenka A, Lanier LL. Natural killer cells, viruses and cancer. Nat Rev Immunol (2001) 1(1):41–9. doi:10.1038/35095564

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Specht A, DeGottardi MQ, Schindler M, Hahn B, Evans DT, Kirchhoff F. Selective downmodulation of HLA-A and -B by Nef alleles from different groups of primate lentiviruses. Virology (2008) 373(1):229–37. doi:10.1016/j.virol.2007.11.019

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lichtfuss GF, Cheng WJ, Farsakoglu Y, Paukovics G, Rajasuriar R, Velayudham P, et al. Virologically suppressed HIV patients show activation of NK cells and persistent innate immune activation. J Immunol (2012) 189(3):1491–9. doi:10.4049/jimmunol.1200458

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol (2004) 22:329–60. doi:10.1146/annurev.immunol.22.012703.104803

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Tu MM, Mahmoud AB, Makrigiannis AP. Licensed and unlicensed NK cells: differential roles in cancer and viral control. Front Immunol (2016) 7:166. doi:10.3389/fimmu.2016.00166

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Kim S, Iizuka K, Aguila HL, Weissman IL, Yokoyama WM. In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc Natl Acad Sci U S A (2000) 97(6):2731–6. doi:10.1073/pnas.050588297

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Street SE, Trapani JA, MacGregor D, Smyth MJ. Suppression of lymphoma and epithelial malignancies effected by interferon gamma. J Exp Med (2002) 196(1):129–34. doi:10.1084/jem.20020063

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Zerafa N, Westwood JA, Cretney E, Mitchell S, Waring P, Iezzi M, et al. Cutting edge: TRAIL deficiency accelerates hematological malignancies. J Immunol (2005) 175(9):5586–90. doi:10.4049/jimmunol.175.9.5586

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Imai K, Matsuyama S, Miyake S, Suga K, Nakachi K. Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population. Lancet (2000) 356(9244):1795–9. doi:10.1016/S0140-6736(00)03231-1

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Fridman WH, Pages F, Sautes-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer (2012) 12(4):298–306. doi:10.1038/nrc3245

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Curigliano G, Criscitiello C, Gelao L, Goldhirsch A. Molecular pathways: human leukocyte antigen G (HLA-G). Clin Cancer Res (2013) 19(20):5564–71. doi:10.1158/1078-0432.CCR-12-3697

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Wu J. NKG2D ligands in cancer immunotherapy: target or not? Austin J Clin Immunol (2014) 1(1):2.

Google Scholar

43. Lin A, Yan WH. HLA-G expression in cancers: roles in immune evasion, metastasis and target for therapy. Mol Med (2015) 21(1):782–91. doi:10.2119/molmed.2015.00083

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Ndhlovu LC, Lopez-Verges S, Barbour JD, Jones RB, Jha AR, Long BR, et al. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood (2012) 119(16):3734–43. doi:10.1182/blood-2011-11-392951

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Han G, Chen G, Shen B, Li Y. Tim-3: an activation marker and activation limiter of innate immune cells. Front Immunol (2013) 4:449. doi:10.3389/fimmu.2013.00449

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Xu L, Huang Y, Tan L, Yu W, Chen D, Lu C, et al. Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK-cell-mediated cytotoxicity in human lung adenocarcinoma. Int Immunopharmacol (2015) 29(2):635–41. doi:10.1016/j.intimp.2015.09.017

CrossRef Full Text | Google Scholar

47. Taborda NA, Hernandez JC, Lajoie J, Juno JA, Kimani J, Rugeles MT, et al. Short communication: low expression of activation and inhibitory molecules on NK cells and CD4(+) T cells is associated with viral control. AIDS Res Hum Retroviruses (2015) 31(6):636–40. doi:10.1089/AID.2014.0325

PubMed Abstract | CrossRef Full Text | Google Scholar

48. da Silva IP, Gallois A, Jimenez-Baranda S, Khan S, Anderson AC, Kuchroo VK, et al. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res (2014) 2(5):410–22. doi:10.1158/2326-6066.CIR-13-0171

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Swart M, Verbrugge I, Beltman JB. Combination approaches with immune-checkpoint blockade in cancer therapy. Front Oncol (2016) 6:233. doi:10.3389/fonc.2016.00233

CrossRef Full Text | Google Scholar

50. Dahlberg CI, Sarhan D, Chrobok M, Duru AD, Alici E. Natural killer cell-based therapies targeting cancer: possible strategies to gain and sustain anti-tumor activity. Front Immunol (2015) 6:605. doi:10.3389/fimmu.2015.00605

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Jones RB, Mueller S, O’Connor R, Rimpel K, Sloan DD, Karel D, et al. A subset of latency-reversing agents expose HIV-infected resting CD4+ T-cells to recognition by cytotoxic T-lymphocytes. PLoS Pathog (2016) 12(4):e1005545. doi:10.1371/journal.ppat.1005545

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Rosario M, Liu B, Kong L, Collins LI, Schneider SE, Chen X, et al. The IL-15-based ALT-803 complex enhances FcgammaRIIIa-triggered NK-cell responses and in vivo clearance of B cell lymphomas. Clin Cancer Res (2016) 22(3):596–608. doi:10.1158/1078-0432.CCR-15-1419

CrossRef Full Text | Google Scholar

53. Seay K, Church C, Zheng JH, Deneroff K, Ochsenbauer C, Kappes JC, et al. In vivo activation of human NK cells by treatment with an interleukin-15 superagonist potently inhibits acute in vivo HIV-1 infection in humanized mice. J Virol (2015) 89(12):6264–74. doi:10.1128/JVI.00563-15

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Scully E, Alter G. NK cells in HIV disease. Curr HIV/AIDS Rep (2016) 13(2):85–94. doi:10.1007/s11904-016-0310-3

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Luteijn R, Sciaranghella G, van Lunzen J, Nolting A, Dugast AS, Ghebremichael MS, et al. Early viral replication in lymph nodes provides HIV with a means by which to escape NK-cell-mediated control. Eur J Immunol (2011) 41(9):2729–40. doi:10.1002/eji.201040886

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Zhou J, Amran FS, Kramski M, Angelovich TA, Elliott J, Hearps AC, et al. An NK-cell population lacking FcRgamma is expanded in chronically infected HIV patients. J Immunol (2015) 194(10):4688–97. doi:10.4049/jimmunol.1402448

CrossRef Full Text | Google Scholar

57. Mavilio D, Lombardo G, Benjamin J, Kim D, Follman D, Marcenaro E, et al. Characterization of CD56-/CD16+ natural killer (NK) cells: a highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A (2005) 102(8):2886–91. doi:10.1073/pnas.0409872102

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med (2012) 366(14):1275–86. doi:10.1056/NEJMoa1113425

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Tomescu C, Chehimi J, Maino VC, Montaner LJ. NK-cell lysis of HIV-1-infected autologous CD4 primary T cells: requirement for IFN-mediated NK activation by plasmacytoid dendritic cells. J Immunol (2007) 179(4):2097–104. doi:10.4049/jimmunol.179.4.2097

CrossRef Full Text | Google Scholar

60. Tomescu C, Mavilio D, Montaner LJ. Lysis of HIV-1-infected autologous CD4+ primary T cells by interferon-alpha-activated NK cells requires NKp46 and NKG2D. AIDS (2015) 29(14):1767–73. doi:10.1097/QAD.0000000000000777

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Tomescu C, Tebas P, Montaner LJ. IFN-alpha augments NK-mediated antibody-dependent cellular cytotoxicity (ADCC) of HIV-1 infected autologous CD4+ T cells regardless of MHC-I downregulation. AIDS (2017) 31(5):613–22. doi:10.1097/QAD.0000000000001380

CrossRef Full Text | Google Scholar

62. Mercier-Bataille D, Sanchez C, Baier C, Le Treut T, Mounier N, Mokhtari S, et al. Expression of activating receptors on natural killer cells from AIDS-related lymphoma patients. AIDS Res Ther (2014) 11:38. doi:10.1186/1742-6405-11-38

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Bruno A, Focaccetti C, Pagani A, Imperatori AS, Spagnoletti M, Rotolo N, et al. The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer. Neoplasia (2013) 15(2):133–42. doi:10.1593/neo.121758

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Li Z, Zhang LJ, Zhang HR, Tian GF, Tian J, Mao XL, et al. Tumor-derived transforming growth factor-beta is critical for tumor progression and evasion from immune surveillance. Asian Pac J Cancer Prev (2014) 15(13):5181–6. doi:10.7314/APJCP.2014.15.13.5181

CrossRef Full Text | Google Scholar

65. Cerdeira AS, Rajakumar A, Royle CM, Lo A, Husain Z, Thadhani RI, et al. Conversion of peripheral blood NK cells to a decidual NK-like phenotype by a cocktail of defined factors. J Immunol (2013) 190(8):3939–48. doi:10.4049/jimmunol.1202582

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Gibellini D, Zauli G, Re MC, Milani D, Furlini G, Caramelli E, et al. Recombinant human immunodeficiency virus type-1 (HIV-1) Tat protein sequentially up-regulates IL-6 and TGF-beta 1 mRNA expression and protein synthesis in peripheral blood monocytes. Br J Haematol (1994) 88(2):261–7. doi:10.1111/j.1365-2141.1994.tb05016.x

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Sawaya BE, Thatikunta P, Denisova L, Brady J, Khalili K, Amini S. Regulation of TNFalpha and TGFbeta-1 gene transcription by HIV-1 Tat in CNS cells. J Neuroimmunol (1998) 87(1–2):33–42. doi:10.1016/S0165-5728(98)00044-7

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Gigante M, Gesualdo L, Ranieri E. TGF-beta: a master switch in tumor immunity. Curr Pharm Des (2012) 18(27):4126–34. doi:10.2174/138161212802430378

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Bruno A, Ferlazzo G, Albini A, Noonan DM. A think tank of TINK/TANKs: tumor-infiltrating/tumor-associated natural killer cells in tumor progression and angiogenesis. J Natl Cancer Inst (2014) 106(8):dju200. doi:10.1093/jnci/dju200

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Liu Q, Sun Y, Rihn S, Nolting A, Tsoukas PN, Jost S, et al. Matrix metalloprotease inhibitors restore impaired NK-cell-mediated antibody-dependent cellular cytotoxicity in human immunodeficiency virus type 1 infection. J Virol (2009) 83(17):8705–12. doi:10.1128/JVI.02666-08

CrossRef Full Text | Google Scholar

71. Azzoni L, Papasavvas E, Chehimi J, Kostman JR, Mounzer K, Ondercin J, et al. Sustained impairment of IFN-gamma secretion in suppressed HIV-infected patients despite mature NK-cell recovery: evidence for a defective reconstitution of innate immunity. J Immunol (2002) 168(11):5764–70. doi:10.4049/jimmunol.168.11.5764

CrossRef Full Text | Google Scholar

72. Goodier MR, Imami N, Moyle G, Gazzard B, Gotch F. Loss of the CD56hiCD16- NK-cell subset and NK-cell interferon-gamma production during antiretroviral therapy for HIV-1: partial recovery by human growth hormone. Clin Exp Immunol (2003) 134(3):470–6. doi:10.1111/j.1365-2249.2003.02329.x

CrossRef Full Text | Google Scholar

73. Chehimi J, Campbell DE, Azzoni L, Bacheller D, Papasavvas E, Jerandi G, et al. Persistent decreases in blood plasmacytoid dendritic cell number and function despite effective highly active antiretroviral therapy and increased blood myeloid dendritic cells in HIV-infected individuals. J Immunol (2002) 168(9):4796–801. doi:10.4049/jimmunol.168.9.4796

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Appay V, Sauce D. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol (2008) 214(2):231–41. doi:10.1002/path.2276

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med (2009) 360(7):692–8. doi:10.1056/NEJMoa0802905

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Ananworanich J, Robb ML. The transient HIV remission in the Mississippi baby: why is this good news? J Int AIDS Soc (2014) 17(1):19859. doi:10.7448/IAS.17.1.19859

CrossRef Full Text | Google Scholar

77. Butler KM, Gavin P, Coughlan S, Rochford A, Donagh SM, Cunningham O, et al. Rapid viral rebound after 4 years of suppressive therapy in a seronegative HIV-1 infected infant treated from birth. Pediatr Infect Dis J (2015) 34:235–40. doi:10.1097/INF.0000000000000570

CrossRef Full Text | Google Scholar

78. Henrich TJ, Hanhauser E, Marty FM, Sirignano MN, Keating S, Lee TH, et al. Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: report of 2 cases. Ann Intern Med (2014) 161(5):319–27. doi:10.7326/M14-1027

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Veluchamy JP, Spanholtz J, Tordoir M, Thijssen VL, Heideman DA, Verheul HM, et al. Combination of NK cells and cetuximab to enhance anti-tumor responses in RAS mutant metastatic colorectal cancer. PLoS One (2016) 11(6):e0157830. doi:10.1371/journal.pone.0157830

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Schoofs T, Klein F, Braunschweig M, Kreider EF, Feldmann A, Nogueira L, et al. HIV-1 therapy with monoclonal antibody 3BNC117 elicits host immune responses against HIV-1. Science (2016) 352(6288):997–1001. doi:10.1126/science.aaf0972

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Byrareddy SN, Arthos J, Cicala C, Villinger F, Ortiz KT, Little D, et al. Sustained virologic control in SIV+ macaques after antiretroviral and alpha4beta7 antibody therapy. Science (2016) 354(6309):197–202. doi:10.1126/science.aag1276

CrossRef Full Text | Google Scholar

82. Felices M, Lenvik TR, Davis ZB, Miller JS, Vallera DA. Generation of BiKEs and TriKEs to improve NK-cell-mediated targeting of tumor cells. Methods Mol Biol (2016) 1441:333–46. doi:10.1007/978-1-4939-3684-7_28

CrossRef Full Text | Google Scholar

83. Gleason MK, Ross JA, Warlick ED, Lund TC, Verneris MR, Wiernik A, et al. CD16xCD33 bispecific killer cell engager (BiKE) activates NK cells against primary MDS and MDSC CD33+ targets. Blood (2014) 123(19):3016–26. doi:10.1182/blood-2013-10-533398

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Gleason MK, Verneris MR, Todhunter DA, Zhang B, McCullar V, Zhou SX, et al. Bispecific and trispecific killer cell engagers directly activate human NK cells through CD16 signaling and induce cytotoxicity and cytokine production. Mol Cancer Ther (2012) 11(12):2674–84. doi:10.1158/1535-7163.MCT-12-0692

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Schmohl JU, Gleason MK, Dougherty PR, Miller JS, Vallera DA. Heterodimeric bispecific single chain variable fragments (scFv) killer engagers (BiKEs) enhance NK-cell activity against CD133+ colorectal cancer cells. Target Oncol (2016) 11(3):353–61. doi:10.1007/s11523-015-0391-8

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Tay SS, Carol H, Biro M. TriKEs and BiKEs join CARs on the cancer immunotherapy highway. Hum Vaccin Immunother (2016) 12(11):2790–6. doi:10.1080/21645515.2016.1198455

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Vallera DA, Felices M, McElmurry R, McCullar V, Zhou X, Schmohl JU, et al. IL15 trispecific killer engagers (TriKE) make natural killer cells specific to CD33+ targets while also inducing persistence, in vivo expansion, and enhanced function. Clin Cancer Res (2016) 22(14):3440–50. doi:10.1158/1078-0432.CCR-15-2710

CrossRef Full Text | Google Scholar

88. Vallera DA, Zhang B, Gleason MK, Oh S, Weiner LM, Kaufman DS, et al. Heterodimeric bispecific single-chain variable-fragment antibodies against EpCAM and CD16 induce effective antibody-dependent cellular cytotoxicity against human carcinoma cells. Cancer Biother Radiopharm (2013) 28(4):274–82. doi:10.1089/cbr.2012.1329

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Reiners KS, Kessler J, Sauer M, Rothe A, Hansen HP, Reusch U, et al. Rescue of impaired NK-cell activity in Hodgkin lymphoma with bispecific antibodies in vitro and in patients. Mol Ther (2013) 21(4):895–903. doi:10.1038/mt.2013.14

CrossRef Full Text | Google Scholar

90. Singer H, Kellner C, Lanig H, Aigner M, Stockmeyer B, Oduncu F, et al. Effective elimination of acute myeloid leukemic cells by recombinant bispecific antibody derivatives directed against CD33 and CD16. J Immunother (2010) 33(6):599–608. doi:10.1097/CJI.0b013e3181dda225

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Wiernik A, Foley B, Zhang B, Verneris MR, Warlick E, Gleason MK, et al. Targeting natural killer cells to acute myeloid leukemia in vitro with a CD16 x 33 bispecific killer cell engager and ADAM17 inhibition. Clin Cancer Res (2013) 19(14):3844–55. doi:10.1158/1078-0432.CCR-13-0505

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Hudak JE, Canham SM, Bertozzi CR. Glycocalyx engineering reveals a Siglec-based mechanism for NK-cell immunoevasion. Nat Chem Biol (2014) 10(1):69–75. doi:10.1038/nchembio.1388

CrossRef Full Text | Google Scholar

93. Xiao H, Woods EC, Vukojicic P, Bertozzi CR. Precision glycocalyx editing as a strategy for cancer immunotherapy. Proc Natl Acad Sci U S A (2016) 113(37):10304–9. doi:10.1073/pnas.1608069113

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Arpinati M, Curti A. Immunotherapy in acute myeloid leukemia. Immunotherapy (2014) 6(1):95–106. doi:10.2217/imt.13.152

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bachanova V, Burns LJ, McKenna DH, Curtsinger J, Panoskaltsis-Mortari A, Lindgren BR, et al. Allogeneic natural killer cells for refractory lymphoma. Cancer Immunol Immunother (2010) 59(11):1739–44. doi:10.1007/s00262-010-0896-z

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Brehm C, Huenecke S, Esser R, Kloess S, Quaiser A, Betz S, et al. Interleukin-2-stimulated natural killer cells are less susceptible to mycophenolate mofetil than non-activated NK cells: possible consequences for immunotherapy. Cancer Immunol Immunother (2014) 63(8):821–33. doi:10.1007/s00262-014-1556-5

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Brehm C, Huenecke S, Quaiser A, Esser R, Bremm M, Kloess S, et al. IL-2 stimulated but not unstimulated NK cells induce selective disappearance of peripheral blood cells: concomitant results to a phase I/II study. PLoS One (2011) 6(11):e27351. doi:10.1371/journal.pone.0027351

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Geller MA, Ivy JJ, Ghebre R, Downs LS Jr, Judson PL, Carson LF, et al. A phase II trial of carboplatin and docetaxel followed by radiotherapy given in a “Sandwich” method for stage III, IV, and recurrent endometrial cancer. Gynecol Oncol (2011) 121(1):112–7. doi:10.1016/j.ygyno.2010.12.338

CrossRef Full Text | Google Scholar

99. Kloess S, Huenecke S, Piechulek D, Esser R, Koch J, Brehm C, et al. IL-2-activated haploidentical NK cells restore NKG2D-mediated NK-cell cytotoxicity in neuroblastoma patients by scavenging of plasma MICA. Eur J Immunol (2010) 40(11):3255–67. doi:10.1002/eji.201040568

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Kottaridis PD, North J, Tsirogianni M, Marden C, Samuel ER, Jide-Banwo S, et al. Two-stage priming of allogeneic natural killer cells for the treatment of patients with acute myeloid leukemia: a phase I trial. PLoS One (2015) 10(6):e0123416. doi:10.1371/journal.pone.0123416

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Penack O, Gentilini C, Fischer L, Asemissen AM, Scheibenbogen C, Thiel E, et al. CD56dimCD16neg cells are responsible for natural cytotoxicity against tumor targets. Leukemia (2005) 19(5):835–40. doi:10.1038/sj.leu.2403704

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Stern M, Passweg JR, Meyer-Monard S, Esser R, Tonn T, Soerensen J, et al. Pre-emptive immunotherapy with purified natural killer cells after haploidentical SCT: a prospective phase II study in two centers. Bone Marrow Transplant (2013) 48(3):433–8. doi:10.1038/bmt.2012.162

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Ullrich E, Koch J, Cerwenka A, Steinle A. New prospects on the NKG2D/NKG2DL system for oncology. Oncoimmunology (2013) 2(10):e26097. doi:10.4161/onci.26097

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Gunther S, Ostheimer C, Stangl S, Specht HM, Mozes P, Jesinghaus M, et al. Correlation of Hsp70 serum levels with gross tumor volume and composition of lymphocyte subpopulations in patients with squamous cell and adeno non-small cell lung cancer. Front Immunol (2015) 6:556. doi:10.3389/fimmu.2015.00556

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Krause SW, Gastpar R, Andreesen R, Gross C, Ullrich H, Thonigs G, et al. Treatment of colon and lung cancer patients with ex vivo heat shock protein 70-peptide-activated, autologous natural killer cells: a clinical phase i trial. Clin Cancer Res (2004) 10(11):3699–707. doi:10.1158/1078-0432.CCR-03-0683

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Specht HM, Ahrens N, Blankenstein C, Duell T, Fietkau R, Gaipl US, et al. Heat shock protein 70 (Hsp70) peptide activated natural killer (NK) cells for the treatment of patients with non-small cell lung cancer (NSCLC) after radiochemotherapy (RCTx) – from preclinical studies to a clinical phase II trial. Front Immunol (2015) 6:162. doi:10.3389/fimmu.2015.00162

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Cartron G, Dacheux L, Salles G, Solal-Celigny P, Bardos P, Colombat P, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene. Blood (2002) 99(3):754–8. doi:10.1182/blood.V99.3.754

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Goede V, Fischer K, Busch R, Engelke A, Eichhorst B, Wendtner CM, et al. Obinutuzumab plus chlorambucil in patients with CLL and coexisting conditions. N Engl J Med (2014) 370(12):1101–10. doi:10.1056/NEJMoa1313984

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Junttila TT, Parsons K, Olsson C, Lu Y, Xin Y, Theriault J, et al. Superior in vivo efficacy of afucosylated trastuzumab in the treatment of HER2-amplified breast cancer. Cancer Res (2010) 70(11):4481–9. doi:10.1158/0008-5472.CAN-09-3704

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Messersmith WA, Ahnen DJ. Targeting EGFR in colorectal cancer. N Engl J Med (2008) 359(17):1834–6. doi:10.1056/NEJMe0806778

CrossRef Full Text | Google Scholar

111. Yu AL, Gilman AL, Ozkaynak MF, London WB, Kreissman SG, Chen HX, et al. Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma. N Engl J Med (2010) 363(14):1324–34. doi:10.1056/NEJMoa0911123

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Benson DM Jr, Bakan CE, Mishra A, Hofmeister CC, Efebera Y, Becknell B, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood (2010) 116(13):2286–94. doi:10.1182/blood-2010-02-271874

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Benson DM Jr, Cohen AD, Jagannath S, Munshi NC, Spitzer G, Hofmeister CC, et al. A phase I trial of the anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma. Clin Cancer Res (2015) 21(18):4055–61. doi:10.1158/1078-0432.CCR-15-0304

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Chan CJ, Martinet L, Gilfillan S, Souza-Fonseca-Guimaraes F, Chow MT, Town L, et al. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol (2014) 15(5):431–8. doi:10.1038/ni.2850

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Gleason MK, Lenvik TR, McCullar V, Felices M, O’Brien MS, Cooley SA, et al. Tim-3 is an inducible human natural killer cell receptor that enhances interferon gamma production in response to galectin-9. Blood (2012) 119(13):3064–72. doi:10.1182/blood-2011-06-360321

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Guo Y, Feng X, Jiang Y, Shi X, Xing X, Liu X, et al. PD1 blockade enhances cytotoxicity of in vitro expanded natural killer cells towards myeloma cells. Oncotarget (2016) 7(30):48360–74. doi:10.18632/oncotarget.10235

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Vey N, Bourhis JH, Boissel N, Bordessoule D, Prebet T, Charbonnier A, et al. A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood (2012) 120(22):4317–23. doi:10.1182/blood-2012-06-437558

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Westin JR, Chu F, Zhang M, Fayad LE, Kwak LW, Fowler N, et al. Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. Lancet Oncol (2014) 15(1):69–77. doi:10.1016/S1470-2045(13)70551-5

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Boissel L, Betancur M, Lu W, Wels WS, Marino T, Van Etten RA, et al. Comparison of mRNA and lentiviral based transfection of natural killer cells with chimeric antigen receptors recognizing lymphoid antigens. Leuk Lymphoma (2012) 53(5):958–65. doi:10.3109/10428194.2011.634048

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Boissel L, Betancur-Boissel M, Lu W, Krause DS, Van Etten RA, Wels WS, et al. Retargeting NK-92 cells by means of CD19- and CD20-specific chimeric antigen receptors compares favorably with antibody-dependent cellular cytotoxicity. Oncoimmunology (2013) 2(10):e26527. doi:10.4161/onci.26527

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Chen X, Han J, Chu J, Zhang L, Zhang J, Chen C, et al. A combinational therapy of EGFR-CAR NK cells and oncolytic herpes simplex virus 1 for breast cancer brain metastases. Oncotarget (2016) 7(19):27764–77. doi:10.18632/oncotarget.8526

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Chu J, Deng Y, Benson DM, He S, Hughes T, Zhang J, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia (2014) 28(4):917–27. doi:10.1038/leu.2013.279

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Esser R, Muller T, Stefes D, Kloess S, Seidel D, Gillies SD, et al. NK cells engineered to express a GD2 -specific antigen receptor display built-in ADCC-like activity against tumour cells of neuroectodermal origin. J Cell Mol Med (2012) 16(3):569–81. doi:10.1111/j.1582-4934.2011.01343.x

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Genssler S, Burger MC, Zhang C, Oelsner S, Mildenberger I, Wagner M, et al. Dual targeting of glioblastoma with chimeric antigen receptor-engineered natural killer cells overcomes heterogeneity of target antigen expression and enhances antitumor activity and survival. Oncoimmunology (2016) 5(4):e1119354. doi:10.1080/2162402X.2015.1119354

CrossRef Full Text | Google Scholar

125. Han J, Chu J, Keung Chan W, Zhang J, Wang Y, Cohen JB, et al. CAR-engineered NK cells targeting wild-type EGFR and EGFRvIII enhance killing of glioblastoma and patient-derived glioblastoma stem cells. Sci Rep (2015) 5:11483. doi:10.1038/srep11483

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Konstantinidis KV, Alici E, Aints A, Christensson B, Ljunggren HG, Dilber MS. Targeting IL-2 to the endoplasmic reticulum confines autocrine growth stimulation to NK-92 cells. Exp Hematol (2005) 33(2):159–64. doi:10.1016/j.exphem.2004.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Liu H, Yang B, Sun T, Lin L, Hu Y, Deng M, et al. Specific growth inhibition of ErbB2 expressing human breast cancer cells by genetically modified NK92 cells. Oncol Rep (2015) 33(1):95–102. doi:10.3892/or.2014.3548

CrossRef Full Text | Google Scholar

128. Miller JS, Soignier Y, Panoskaltsis-Mortari A, McNearney SA, Yun GH, Fautsch SK, et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood (2005) 105(8):3051–7. doi:10.1182/blood-2004-07-2974

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Muller T, Uherek C, Maki G, Chow KU, Schimpf A, Klingemann HG, et al. Expression of a CD20-specific chimeric antigen receptor enhances cytotoxic activity of NK cells and overcomes NK-resistance of lymphoma and leukemia cells. Cancer Immunol Immunother (2008) 57(3):411–23. doi:10.1007/s00262-007-0383-3

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Nagashima S, Mailliard R, Kashii Y, Reichert TE, Herberman RB, Robbins P, et al. Stable transduction of the interleukin-2 gene into human natural killer cell lines and their phenotypic and functional characterization in vitro and in vivo. Blood (1998) 91(10):3850–61.

PubMed Abstract | Google Scholar

131. Ni Z, Knorr DA, Kaufman DS. Hematopoietic and nature killer cell development from human pluripotent stem cells. Methods Mol Biol (2013) 1029:33–41. doi:10.1007/978-1-62703-478-4_3

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Oelsner S, Wagner J, Friede ME, Pfirrmann V, Genssler S, Rettinger E, et al. Chimeric antigen receptor-engineered cytokine-induced killer cells overcome treatment resistance of pre-B-cell acute lymphoblastic leukemia and enhance survival. Int J Cancer (2016) 139(8):1799–809. doi:10.1002/ijc.30217

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Romanski A, Uherek C, Bug G, Seifried E, Klingemann H, Wels WS, et al. CD19-CAR engineered NK-92 cells are sufficient to overcome NK-cell resistance in B-cell malignancies. J Cell Mol Med (2016) 20(7):1287–94. doi:10.1111/jcmm.12810

CrossRef Full Text | Google Scholar

134. Schirrmann T, Pecher G. Human natural killer cell line modified with a chimeric immunoglobulin T-cell receptor gene leads to tumor growth inhibition in vivo. Cancer Gene Ther (2002) 9(4):390–8. doi:10.1038/sj.cgt.7700453

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Schonfeld K, Sahm C, Zhang C, Naundorf S, Brendel C, Odendahl M, et al. Selective inhibition of tumor growth by clonal NK cells expressing an ErbB2/HER2-specific chimeric antigen receptor. Mol Ther (2015) 23(2):330–8. doi:10.1038/mt.2014.219

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Tam YK, Maki G, Miyagawa B, Hennemann B, Tonn T, Klingemann HG. Characterization of genetically altered, interleukin 2-independent natural killer cell lines suitable for adoptive cellular immunotherapy. Hum Gene Ther (1999) 10(8):1359–73. doi:10.1089/10430349950018030

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Uherek C, Tonn T, Uherek B, Becker S, Schnierle B, Klingemann HG, et al. Retargeting of natural killer-cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood (2002) 100(4):1265–73.

PubMed Abstract | Google Scholar

138. Woll PS, Grzywacz B, Tian X, Marcus RK, Knorr DA, Verneris MR, et al. Human embryonic stem cells differentiate into a homogeneous population of natural killer cells with potent in vivo antitumor activity. Blood (2009) 113(24):6094–101. doi:10.1182/blood-2008-06-165225

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Yang B, Liu H, Shi W, Wang Z, Sun S, Zhang G, et al. Blocking transforming growth factor-beta signaling pathway augments antitumor effect of adoptive NK-92 cell therapy. Int Immunopharmacol (2013) 17(2):198–204. doi:10.1016/j.intimp.2013.06.003

CrossRef Full Text | Google Scholar

140. Zhang J, Sun R, Wei H, Zhang J, Tian Z. Characterization of interleukin-15 gene-modified human natural killer cells: implications for adoptive cellular immunotherapy. Haematologica (2004) 89(3):338–47.

PubMed Abstract | Google Scholar

141. Zhang J, Sun R, Wei H, Zhang J, Tian Z. Characterization of stem cell factor gene-modified human natural killer cell line, NK-92 cells: implication in NK-cell-based adoptive cellular immunotherapy. Oncol Rep (2004) 11(5):1097–106. doi:10.3892/or.11.5.1097

CrossRef Full Text | Google Scholar

142. Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol (2014) 32(4):347–55. doi:10.1038/nbt.2842

CrossRef Full Text | Google Scholar

143. Garrido C, Spivak AM, Soriano-Sarabia N, Checkley MA, Barker E, Karn J, et al. HIV latency-reversing agents have diverse effects on natural killer cell function. Front Immunol (2016) 7:356. doi:10.3389/fimmu.2016.00356

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: HIV, cancer, natural killer cells, ADCC, antiretroviral therapy, lymphoma, eradication and control

Citation: Leal FE, Premeaux TA, Abdel-Mohsen M and Ndhlovu LC (2017) Role of Natural Killer Cells in HIV-Associated Malignancies. Front. Immunol. 8:315. doi: 10.3389/fimmu.2017.00315

Received: 16 December 2016; Accepted: 06 March 2017;
Published: 21 March 2017

Edited by:

Sandra Laurence Lopez-Verges, Instituto Conmemorativo Gorgas de Estudios de la Salud, Panama

Reviewed by:

Roland Jacobs, Hannover Medical School, Germany
Amedeo Amedei, University of Florence, Italy

Copyright: © 2017 Leal, Premeaux, Abdel-Mohsen and Ndhlovu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Lishomwa C. Ndhlovu, lndhlovu@hawaii.edu

Present address: Mohamed Abdel-Mohsen, The Wistar Institute, Philadelphia, PA, USA

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