Next Article in Journal
S2 Peptide-Conjugated SARS-CoV-2 Virus-like Particles Provide Broad Protection against SARS-CoV-2 Variants of Concern
Previous Article in Journal
An Overview of the Strategies to Boost SARS-CoV-2-Specific Immunity in People with Inborn Errors of Immunity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas

1
Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
2
Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
3
Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
4
The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
5
Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
6
The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
*
Author to whom correspondence should be addressed.
Vaccines 2024, 12(6), 677; https://doi.org/10.3390/vaccines12060677
Submission received: 19 April 2024 / Revised: 5 June 2024 / Accepted: 14 June 2024 / Published: 18 June 2024
(This article belongs to the Section Cancer Vaccines and Immunotherapy)

Abstract

:
High-grade serous ovarian cancers (HGSOCs) likely consist of poorly differentiated stem-like cells (PDSLCs) and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs, contributing to disease progression and tumor relapse. Primary NK cells are known to effectively lyse PDSLCs, but they exhibit low or minimal cytotoxic potential against well-differentiated tumors. We have introduced and discussed the characteristics of super-charged NK (sNK) cells in this review. sNK cells, in comparison to primary NK cells, exhibit a significantly higher capability for the direct killing of both PDSLCs and well-differentiated tumors. In addition, sNK cells secrete significantly higher levels of cytokines, especially those known to induce the differentiation of tumors. In addition, we propose that a combination of sNK and chemotherapy could be one of the most effective strategies to eliminate the heterogeneous population of ovarian tumors; sNK cells can lyse both PDSLCs and well-differentiated tumors, induce the differentiation of PDSLCs, and could be used in combination with chemotherapy to target both well-differentiated and NK-induced differentiated tumors.

1. Introduction and Background: Ovarian Cancer

Ovarian carcinomas are the second most common cause of gynecological malignancies-related death and are composed of a diverse group of tumors based on histological, molecular, and genetic factors [1]. In total, 70% are high-grade serous ovarian cancers (HGSOCs), 10% are endometrioid, 10% is clear cells, 3% mucinous, and approximately 5% low-grade serous carcinoma [1,2,3,4,5]. HGSOCs arise from the epithelium of the distal fallopian tube, are typically diagnosed at an advanced stage, and are responsible for the majority of ovarian cancer-related deaths [2,3,6,7]. The standard approach of treating HGSOCs is surgical intervention combined with platinum-based chemotherapy [8], which initially yields positive responses in many patients [9]. Despite advances in treatment, the prognosis of HGSOC remains poor, and the majority of treated cases relapse within a few years of diagnosis resulting in a 5-year survival rate lower than 40% [7]. Thus, the high recurrence rate and chemoresistance development highlight the urgent need for novel treatment strategies to improve patient prognosis [10]. Recently, the FDA approved mirvetuximab soravtansine as a novel therapeutic alternative for a subset of platinum-resistant patients with high levels of folate receptor alpha expression within the tumor [11,12]. While this therapy provides an alternative treatment for this subpopulation, the observed median post-treatment progression-free survival in cancer patients was 4–5.6 months [11,13], underscoring the urgent need to develop innovative treatment approaches.
Immunotherapy has emerged as a promising approach for cancer treatment, particularly for HGSOC. Natural killer (NK) cells, a type of cytotoxic lymphocyte, play a crucial role in the innate immune system and have demonstrated potential in targeting cancer cells [14,15,16]. This review explores the potential of NK cell-based immunotherapy in targeting HGSOC, focusing on the characteristics and advantages of supercharged NK (sNK) cells.

2. Heterogeneity and Mechanism of Chemoresistance in HGSOC

The complexity of treating HGSOCs is exacerbated by the tumor’s heterogeneity [17] and the absence of clearly targetable driver mutations [18]. Studies have demonstrated that most of the mutations in primary tumors persist post chemotherapy suggesting that most clones are chemoresistant [18,19]. It was found that pre-treatment subclones of the tumor population could undergo expansion during chemotherapy or platinum-based therapy resulting in tumor relapse [20,21]. Several studies were performed to investigate the mechanism of chemoresistance in HGSOC. Notably, platinum-based therapies, while effective in eradicating most tumor cells, likely leave behind a population of poorly differentiated stem-like cells implicated in disease progression and tumor relapse [22,23,24,25]. The identification and targeting of these tumor stem-like cells have been explored in various studies and found increased expression levels of cell surface markers like CD24, CD44, CD133, CD117, elevated ALDH activity, and increased levels of pluripotency-related transcription factors such as Nanog, Oct3/4, and Sox2 [26,27,28,29,30,31]. Indeed, the increased expression of pluripotency-related transcription factors is seen in recurrent HGSOC tumor samples compared to chemo-naive ones [32].
It is now well-known that the tumor tissues consist of diverse cellular components including cancer cells and stromal cells [5]. IL-6 was found to induce chemoresistance in ovarian cancer, and mesenchymal stromal cells, particularly cancer-associated fibroblasts (CAFs), were found to be the main source of IL-6 secretion in ovarian cancer [33]. It was found that CCL2 and CCL5 secreted by CAFs could stimulate IL-6 secretion in ovarian cancer cells ultimately resulting in chemoresistance in tumors [34]. Several downstream mechanisms including PYK2, Ras/MEK/ERK, PI3K/Akt, and JAK/STAT3 signaling play a role in IL-6-mediated chemoresistance in ovarian cancers [33,34,35]. Leung et al. demonstrated that a crosstalk signal of CAFs and endothelial cells could lead to chemoresistance in ovarian cancer via regulation of the lipoma preferred partner (LPP) gene in endothelial cells [36]. Stromal cell-derived midkine was found to associated with chemoresistance in ovarian cancer due to increased expression levels of IncRNA ANRIL [37]. Gonzalez et al. performed in-depth single-cell phenotypic characterization of HGSOC using multiparametric mass cytometry (cyTOF) and found that HGSOC expressing higher levels of vimentin, cMyc, and HE4 in the presence of a low expression level of E-cadherin was carboplatin resistant [38]. It was found that an increased protein expression level of SUSD2 (Notch3-regulating gene) in HGSOC promotes epithelial–mesenchymal transition (EMT), the metastatic capacity of malignant cells, and chemoresistance [39]. Higher expression of oncogene KDM5A was found in ovarian cancer tissues and is associated with cancer cell proliferation, EMT, metastasis, and chemoresistance [40]. Expression of CD44V8-10 (CD44 variant containing exons v8-10) was found associated with chemoresistance and a poor prognosis in ovarian cancer [41].
While there may not be consensus on the exact expression profile of the HGSOC stem-like cells, most in this field believe they exist and may be responsible for tumor relapse and chemoresistance. Henceforth, we will refer to these cells as poorly differentiated stem-like cells (PD-SLCs) (Figure 1). In addition to chemotherapy resistance, the challenge of immune evasion by PD-SLCs within HGSOC tumors has proven to be a large hurdle to overcome as these cells employ mechanisms that reduce immunogenicity such as the altered expression of surface antigens to escape T-cell recognition [42]. However, some of these cells remain susceptible to natural killer (NK) cell-mediated cytotoxicity due to their low MHC-class I expression, allowing NK cells to kill these cells through various mechanisms like the release of cytotoxic molecules, cytokines, or chemokines [43]. This phenomenon has opened a further investigation into using NK cell-based therapies to target PD-SLCs in HGSOC, a malignancy that can harbor tumor cells with the down-modulation of MHC-class I [44].

3. Immunosuppression in Ovarian Cancer Patients: Rationale for Increased Metastasis

3.1. Immunosuppressive Nature of Ovarian Tumors

To understand the mechanisms of tumor progression, we focused on the role of the tumor cells to mediate immunosuppressive effects within the tumor microenvironment. A prospective cohort study conducted on ovarian cancer patients has shown that increased ascites are associated with a poor prognosis and increased immunosuppression [45]. Intratumoral T cells (T cells with tumor-cell islets) were found to positively contribute to a better prognosis in advanced ovarian carcinoma [46]. Decreased intratumoral T cells were found to be associated with the up-regulation of vascular endothelial growth factor [46]. Ovarian carcinoma ascites-derived tumor-associated T and NK cells were found to exhibit the defective expression and function of signaling proteins including decreased expression levels of TcR-zeta chains and p56 (lck), and reduced gene expression levels of IFN-γ, IL-2, and IL-4 [47]. Although ovarian cancer patients exhibit higher concentrations of NK cells in ascites fluid as compared to peripheral blood, these NK cells lack function [48,49]. Ascites-derived NK cells were found to express low surface expression of CD16, NKp30, and NKG2D, and were less capable of expanding, mediating cytotoxicity, and secreting cytokines in comparison to peripheral blood-derived NK cells [50,51,52]. Lower levels of NK cell numbers were seen in the peripheral blood of ovarian cancer patients [49,53]. The study conducted on the lymphocytes collected from the peritoneal cavity of ovarian cancer patients exhibited dysfunctional lymphocyte-mediated direct and ADCC cytotoxic functions contributing to the spread and proliferation of tumor cells in the peritoneal cavity [48,54].

3.2. Markers Associated with Better Prognosis

It was found that the overexpression of the endothelin B receptor (ETBR) is associated with a lack of tumor-infiltrating lymphocytes (TILs) and a shortened life span in ovarian cancer patients [55]. Although CD103+ TILs comprising CD8+ T cells and CD56+ NK cells were found in most ovarian cancer types, they were found to be the most abundant in HGSOCs and were associated with patient survival [56]. CD103 was found to serve as a useful marker to enrich the most beneficial subsets for immunotherapy [56]. Higher percentages, function, and IL-15-mediated activation of ascites-derived NK cells were found to be associated with a better prognosis in ovarian cancer patients [57]. The IL-15 super-agonist complex was found to up-regulate NK cell-mediated cytotoxicity against ovarian cancer cell lines in both in vitro and in vivo studies [58].

3.3. Immune Cells Associated with Better Prognosis

A study conducted on the interaction between dendritic cells and T cells has shown that tumor-associated plasmacytoid dendritic cells were found to contribute to tumor immunosuppressive networks in ovarian cancer patients [59]. Tumor-associated T-reg cells were found to be positive for the expression of NKG2D ligand surface expression and were found to be killed by T cells expressing chimeric NKG2D (chNKG2D) receptors due to receptor–ligand interaction in a perforin-dependent manner [60]. Studies have shown that ovarian tumor cells and tumor microenvironmental macrophages produce the chemokine CCL22, responsible for the preferential recruitment and accumulation of T-regs in tumors and in ascites [61]. Blocking the function or migration of T-reg to the tumor microenvironment could fight the battle against ovarian cancer [61]. A higher number of intraepithelial CD8+ TILs and an increased ratio of CD8+ vs. T-reg cells were found to be associated with a better prognosis of epithelial ovarian cancer [62].

3.4. Limitations and Challenge in HGSOC Treatments

While NK cell-based immunotherapy holds significant promise for HGSOC treatment, it is essential to acknowledge its potential limitations and challenges. One major challenge is the immunosuppressive tumor microenvironment, which can hinder NK cell function and efficacy. Additionally, the heterogeneity of HGSOC tumors, with varying expression of NK cell-activating ligands and MHC-class I molecules, poses a challenge for targeting all tumor cell populations by primary activated NK cells.

3.5. Future Strategies to Overcome the Challenges of HGSOC Treatments

Future research should focus on developing strategies to overcome these challenges. This includes investigating methods to enhance NK cell infiltration and function within the tumor microenvironment, as well as exploring combination therapies with other immunomodulatory agents or targeted therapies. Additionally, further research is needed to identify and target specific tumor cell populations that may be resistant to primary activated NK cell-mediated cytotoxicity. We have designed and implemented a strategy to use supercharged NK cells as an effective strategy to target the heterogenous nature of ovarian tumors which primary activated NK cells are not capable of targeting.

4. Immunotherapies of Ovarian Cancer

Cancer immunotherapies have gained popularity due to their increased effectiveness as a cancer therapy. Humanized monoclonal antibody bevacizumab (anti-vascular endothelial growth factor) adjuvant therapy combined with chemotherapy has been approved for advanced primary and recurrent tumors resulting in a moderate improvement in progression-free survival [63,64,65]. The anti-tumor responses of antibodies against PD1/PDL1 were investigated in ovarian cancer. Monotherapies with nivolumab [66] or pembrolizumab [67] were found to be effective only in a small fraction of ovarian cancer patients. While the combination of nivolumab and ipilimumab therapy was found superior to nivolumab alone, its efficacy remains limited [68]. T cells expressing chimeric NKG2D (chNKG2D) receptors were found to contribute to long-term tumor-free survival in ovarian cancer mouse models [60]. The p53 synthetic long peptide (p53-SLP) was found to be a safe vaccine to induce T-cell responses in ovarian cancer patients [69,70]. Mesothelin-induced CAR-T prolonged survival in an ovarian cancer mouse model [71].
Ovarian tumors have been shown to be targeted by NK cell-mediated killing in several in vitro and in vivo assays [58,72,73,74,75,76], including against 3D ovarian tumor spheroids [75]. It was observed that IP injection of iPSC-derived NK cells and/or peripheral blood-derived NK cells led to high levels of circulating NK cells and the eradication of intraperitoneal ovarian cancer in xenograft mice model [72,73]. Ovarian patient peripheral blood and ascites-derived ex vivo expanded NK cells were found to be cytotoxic against autologous primary ovarian tumor cell lines [76]. Also, allogeneic ex vivo expanded NK cell treatment resulted in a reduced tumor burden and tumor metastasis in mouse models implanted with patient-derived ovarian tumors [77,78]. Adoptive transfer of haploidentical NK cells in combination with chemotherapy was found to be effective in treating ovarian cancer due to the in vivo increased survival and expansion of NK cells; however, strategies to augment in vivo NK cell persistence and expansion need to be explored further [79].

5. The Role of MHC-Class I and II Expression on Ovarian Tumors and Their Susceptibility to NK Cell-Mediated Effects

The mechanisms that govern NK activation are still under investigation. However, increased NK cell function has been seen upon the deletion or decrease in many cellular genes in tumors [80]. Most important, the down-modulation of NF-κB and CD44 in tumors induced a significant increase in both cytotoxic activity and the secretion levels of IFN-γ in NK cells in vitro, and induced inflammation and auto-immunity in vivo [14,80,81,82,83]. In our gene knock-down studies, the down-modulation of MHC-class I expression in both transformed and non-transformed healthy cells was found to be one of the underlying mechanisms to induce activation in NK cells [14,80,82,83]. Recent discoveries suggest that NK cell activation is much more complex than we have previously envisioned; it involves many genes/pathways. For example, it was found in mouse studies that DAP10/DAP12 knock-outs resulted in hyper-responsiveness in NK cells [84]. Thus, increased responsiveness of NK cells when key cellular genes were knocked out or knocked down in interacting cells/tumors may point to the fundamental function of NK cells in targeting cells that lose the ability to differentiate optimally, and that the degree of differentiation of the cells is likely the key in regulating NK cell expansion and function.
Using several cancer cell lines, we have previously demonstrated that PDSLCs exhibit lower MHC-class I expression, whereas well-differentiated tumors exhibit much higher expression of surface MHC-class I [14,85,86]. To understand the role of MHC-class I in susceptibility to NK cell-mediated cytotoxicity against ovarian tumors, we have previously used seven ovarian tumor cell lines: OVCAR3 [87,88,89,90], OVCAR4 [91,92,93], OVCAR8 [94,95,96], SKOV3 [77,97,98], Kuramochi [94,99], CaOV3 [77,100,101,102], and OAW28 [86,103,104]. There was a great correlation between the levels of MHC-class I expression and targeting by NK cells. Both OVCAR8 and CAOV3 had minimal expression of MHC-class I and were highly killed by NK cells when compared to the other tumor lines [44]. The down-modulation or loss of MHC-class I and class II has been reported in ovarian carcinoma [105,106,107,108,109] and is one of the factors contributing to escape by the immune system [110]. Increased expression levels of MHC-class I and class II in ovarian cancer correlate with increased numbers of tumor-infiltrating lymphocytes, an increased response to PD1/PDL1 therapy, better prognosis, and the prolonged life of cancer patients [108,109,111,112].
Considering that there is significant down-modulation of surface receptors when cellular genes are decreased or deleted, it is likely that NK cells sense the concentration of receptors on the surface of the cells and react accordingly. The higher the loss of receptors, the more activation of NK cells is expected. In contrast, the rise in surface receptors shuts down the primary NK function mostly known to be due to the down-modulation of MHC-class I. Other receptors are also clearly important in the process. This aspect of NK cell activation requires future investigations.

6. Supercharged NK Cells as an Effective Strategy to Eliminate Aggressive Ovarian Tumors

NK cells target tumors via direct cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC) or can indirectly regulate the functions of other immune effectors through their secreted inflammatory cytokines and chemokines [113,114,115]. Increased NK cell activity in peripheral blood and/or NK cell infiltration of tumor tissue are associated with a better prognosis in cancer patients [116,117,118,119]. NK cell-based cancer immunotherapies are found to be effective against several solid tumors [79,120,121,122,123,124]. Studies have shown an increased sensitivity of HGSOC tumor cells to NK cell-mediated cytotoxicity [72,73,79,125] and the ubiquitous expression of NK ligands on primary HGSOC tumors [32]. Our data also reveal higher NK-mediated cytotoxicity against HGSOC cell lines expressing lower MHC-class I levels [44]. Despite the potential of NK cells in targeting HGSOC, their effectiveness is limited by several factors: (i) a restricted number of NK cells in the tumor, (ii) dysfunctional endogenous NK cells among ovarian cancer patients, (iii) the short lifespan of NK cells in vivo, (iv) difficulties in NK cell infiltration to the tumor site, particularly in solid tumors, and (v) functional impairments in the tumor microenvironment [43,126,127].
To overcome the challenges mentioned above regarding NK-cell therapies, our laboratory has developed the technology to up-regulate the proliferative, effector, and cytotoxic function of NK cells. This technique involves the activation of peripheral blood-derived NK cells with a combination of IL-2 and anti-CD16 mAbs, and co-culturing of activated NK cells with osteoclasts (OCs) as feeder cells in the presence of probiotics sAJ2, this process leads to expansion and highly functional activation of NK cells [103,128,129]. Due to their superior anti-cancer activity, these cells were named supercharged NK (sNK) cells [15,16,86,103,128,129,130,131,132,133,134,135,136] (Table 1 and Figure 2). sNK cells were found to be highly effective against tumors in preclinical models [15,16,86,103,128,129,130,131,132,133,134,135,136]. The rationale for using OCs for this methodology was the secretion of a wide range of cytokines and chemokines including IL-12, IL-15, IFN-a, and IL-18 by OCs, which are known to increase NK function. In addition, OCs express the important NK-activating ligands MICA/B and ULBPs [137,138]. The probiotic sAJ2 is a combination of Gram-positive probiotic bacteria strains: Streptococcus thermophiles, Bifidobacterium longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus casei, and Lactobacillus bulgaricus. The rationale for using probiotic sAJ2 treatments was to increase the cytokines secreted by NK cells including IFN-γ which could facilitate the signals required for NK cell expansion [85,86,128,135,139]. Therefore, combining both probiotics and OCs led to the induction of signals participating in the expansion, survival, and functional activation of NK cells (Table 1 and Figure 2).
We have previously demonstrated that sNK cells exhibit a significantly higher capability for cytokine secretion and cytotoxic function against several cancers (including ovarian cancers) in comparison to activated primary NK cells [128,130,140]. Both the cytokine secretion and cytotoxic activity of sNK cells were maintained for over 30 days (donor dependent: average 27–36 days), as opposed to the 5–7-day duration observed in primary activated NK cells [128,130,140]. Thus far, it is very well established that primary IL-2-activated NK cells typically target PDSLCs and exhibit a slight/low killing potential against well-differentiated tumors. This pattern did not apply to the sNK-induced killing of tumors because sNK cells were found to target both PDSLCs and well-differentiated tumors. We observed that sNK cells induced a significantly greater killing of differentiated tumors when compared to primary activated NK cells [44,128,129,132,135,140]. In addition, IFN-γ secreted by sNK cells induced significantly higher differentiation of PDSLCs when compared to primary activated NK cells [128,130] (Table 1). Although differentiated tumors become resistant to primary activated NK cell-mediated cytotoxicity, they remain susceptible to sNK cell-mediated cytotoxicity and chemotherapeutic drugs [14,130,132], and such an effect was also seen in ovarian cancer cell lines [44].
In our recent study, we found that sNK cells were the only NK cells that effectively lysed both well-differentiated and poorly differentiated oral and ovarian tumors, as evidenced in both the normalized cell index and at the level of cytolysis, whereas primary activated NK cells could only lyse poorly differentiated tumors. In all cases, sNK cells greatly lysed the tumor cells and no visible tumor could be visualized. Therefore, sNK cells behave not only as NK cells but also as T cells, since they are capable of lysing MHC-class I-bearing well-differentiated tumors.
We are now in the process of investigating the efficacy of sNK cells in ovarian tumor-bearing humanized-BLT mice. In our previous studies, we performed one intravenous injection of sNK cells in oral [130,135], pancreatic [86,130], and melanoma [manuscript in prep] tumors implanted humanized-BLT mice, and mice were monitored for 4–5 weeks. Mice received sNK cells therapy had an increased lifespan and decreased tumor mass in comparison to mice receiving no treatment [86,130,135]. Tumors dissected from the sNK cell-treated group expressed higher expression levels of human CD45, MHC-class I, CD54, and B7H1 [86,130,135]. These results suggest that sNK cells increased tumor immune cell infiltration and induced in vivo tumor differentiation. In addition, the restored cytotoxic function and cytokine secretion of immune cells isolated from the spleen, peripheral blood, gingiva, pancreas, and bone marrow of sNK cell-treated tumor-bearing hu-BLT mice were observed [86,130,135].

7. Clinical Trials of NK Cell-Based Ovarian Cancer Therapeutics

Several immunotherapeutic clinical trials have demonstrated the efficacy of immunotherapy as a treatment modality for cancers [141,142,143,144,145]. NK cell-based immunotherapies have demonstrated benefits in clinical trials to treat several cancers including ovarian cancer [123,146,147,148,149,150]. Allogeneic NK cell transfer is safe and feasible because NK cells do not require HLA matching [151]. Geller et al. (ClinicalTrials.gov. identifier: NCT01105650) enrolled 14 ovarian cancer patients, which were infused with haploidentical IL-2 activated NK cells followed by SC IL-2 infusion three times per week for fourteen days, after which NK cell expansion was detected in the peripheral blood of patients [79]. Xie et al. documented a case report of ovarian cancer patient receiving ex vivo expanded NK cells every two weeks for a total of six infusions; this approach resulted in a significant reduction in tumor mass and prolonged patient survival in the presence of minimal adverse effects [78]. A summary of NK cell-based clinical trials for ovarian cancer is listed in Table 2. In total, 6 out of 14 patients enrolled in the clinical trial NCT01105650 were found to have progression-free survival at one year. For other listed trials, evaluations such as safety, progression-free survival, or overall survival are still under investigation.
In addition, we have recently initiated clinical trials using sNK cells. The results from our first two patients exhibited an unparalleled safety profile and initial positive results. We will be continuously monitoring these patients to obtain data for their efficacy.

8. Conclusions

Both primary activated NK cells and sNK cells play a crucial role in the direct killing and differentiation of ovarian tumors. However, sNK cells are more potent, ultimately preventing tumor establishment by the specific recognition, selection, and differentiation of tumors. Preclinical investigations and clinical trials (ongoing and completed) have demonstrated the promising anti-cancer effects of primary activated NK cell-based immunotherapy. Because sNK cells induce significant direct killing and/or differentiation of PDSLCs, and chemotherapy selectively targets differentiated tumors, a combination of both sNK and chemotherapy could be the most effective strategy to eliminate the heterogeneous population of ovarian tumors (Figure 3). Current work in progress will provide further information needed to establish efficacy based on the tumor site, stage of cancer, and patient-related factors, and add more information regarding the optimal dosage of NK cell infusion in combination with other therapeutics. The work presented in this review suggests NK cell-based therapy will be an important part of the armamentarium of ovarian cancer therapeutics.

Author Contributions

A.J. and S.M. were the principal investigators, obtained the funding, designed the study, and wrote the manuscript along with K.K. K.K. performed the literature search and wrote the manuscript. J.S. prepared figures and participated in writing and editing the manuscript. All figures created with BioRender.com on 30 April 2024. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by a UCLA Eli and Edythe Board Center of Regenerative Medicine and Stem Cell Research Award, funded by the Binder Foundation awarded to S.M. and A.J.

Conflicts of Interest

The authors declare no conflicts of interest.

List of Abbreviations

HGSOCsHigh-grade serous ovarian cancers
PDSLCsPoorly differentiated stem-like cells
NK cellsNatural killer cells
sNK cellsSupercharged NK cells
IFN-γInterferon-gamma
CAFsCancer-associated fibroblasts
IL-6Interleukin 6
LPPLipoma preferred partner
OCsOsteoclasts
EMTEpithelial–mesenchymal transition
ETBREndothelin B receptor
TILsTumor-infiltrating lymphocytes
MHC-class IMajor histocompatibility complex molecule class I
ADCCAntibody-dependent cellular cytotoxicity (ADCC)

References

  1. Siegel, R.L.; Giaquinto, A.N.; Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 2024, 74, 12–49. [Google Scholar] [CrossRef] [PubMed]
  2. Prat, J. Ovarian carcinomas: Five distinct diseases with different origins, genetic alterations, and clinicopathological features. Virchows Arch. 2012, 460, 237–249. [Google Scholar] [CrossRef] [PubMed]
  3. McCluggage, W.G. Morphological subtypes of ovarian carcinoma: A review with emphasis on new developments and pathogenesis. Pathology 2011, 43, 420–432. [Google Scholar] [CrossRef] [PubMed]
  4. Kurman, R.J.; Shih Ie, M. The origin and pathogenesis of epithelial ovarian cancer: A proposed unifying theory. Am. J. Surg. Pathol. 2010, 34, 433–443. [Google Scholar] [CrossRef]
  5. Kim, S.; Kim, B.; Song, Y.S. Ascites modulates cancer cell behavior, contributing to tumor heterogeneity in ovarian cancer. Cancer Sci. 2016, 107, 1173–1178. [Google Scholar] [CrossRef] [PubMed]
  6. Bell, D.; Berchuck, A.; Birrer, M.; Chien, J.; Cramer, D.W.; Dao, F.; Dhir, R.; DiSaia, P.; Gabra, H.; Glenn, P.; et al. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef] [PubMed]
  7. Lahtinen, A.; Lavikka, K.; Virtanen, A.; Li, Y.; Jamalzadeh, S.; Skorda, A.; Lauridsen, A.R.; Zhang, K.; Marchi, G.; Isoviita, V.M.; et al. Evolutionary states and trajectories characterized by distinct pathways stratify patients with ovarian high grade serous carcinoma. Cancer Cell 2023, 41, 1103–1117.e1112. [Google Scholar] [CrossRef] [PubMed]
  8. Kurnit, K.C.; Fleming, G.F.; Lengyel, E. Updates and New Options in Advanced Epithelial Ovarian Cancer Treatment. Obs. Gynecol. 2021, 137, 108–121. [Google Scholar] [CrossRef]
  9. Kim, A.; Ueda, Y.; Naka, T.; Enomoto, T. Therapeutic strategies in epithelial ovarian cancer. J. Exp. Clin. Cancer Res. 2012, 31, 14. [Google Scholar] [CrossRef] [PubMed]
  10. Baert, T.; Ferrero, A.; Sehouli, J.; O’Donnell, D.M.; Gonzalez-Martin, A.; Joly, F.; van der Velden, J.; Blecharz, P.; Tan, D.S.P.; Querleu, D.; et al. The systemic treatment of recurrent ovarian cancer revisited. Ann. Oncol. 2021, 32, 710–725. [Google Scholar] [CrossRef] [PubMed]
  11. Matulonis, U.A.; Lorusso, D.; Oaknin, A.; Pignata, S.; Dean, A.; Denys, H.; Colombo, N.; Van Gorp, T.; Konner, J.A.; Marin, M.R.; et al. Efficacy and Safety of Mirvetuximab Soravtansine in Patients With Platinum-Resistant Ovarian Cancer with High Folate Receptor Alpha Expression: Results From the SORAYA Study. J. Clin. Oncol. 2023, 41, 2436–2445. [Google Scholar] [CrossRef] [PubMed]
  12. Dilawari, A.; Shah, M.; Ison, G.; Gittleman, H.; Fiero, M.H.; Shah, A.; Hamed, S.S.; Qiu, J.; Yu, J.; Manheng, W.; et al. FDA Approval Summary: Mirvetuximab Soravtansine-Gynx for FRα-Positive, Platinum-Resistant Ovarian Cancer. Clin. Cancer Res. 2023, 29, 3835–3840. [Google Scholar] [CrossRef] [PubMed]
  13. Moore, K.N.; Angelergues, A.; Konecny, G.E.; García, Y.; Banerjee, S.; Lorusso, D.; Lee, J.Y.; Moroney, J.W.; Colombo, N.; Roszak, A.; et al. Mirvetuximab Soravtansine in FRα-Positive, Platinum-Resistant Ovarian Cancer. N. Engl. J. Med. 2023, 389, 2162–2174. [Google Scholar] [CrossRef] [PubMed]
  14. Kozlowska, A.K.; Topchyan, P.; Kaur, K.; Tseng, H.C.; Teruel, A.; Hiraga, T.; Jewett, A. Differentiation by NK cells is a prerequisite for effective targeting of cancer stem cells/poorly differentiated tumors by chemopreventive and chemotherapeutic drugs. J. Cancer 2017, 8, 537–554. [Google Scholar] [CrossRef] [PubMed]
  15. Jewett, A.; Kos, J.; Fong, Y.; Ko, M.W.; Safaei, T.; Perišić Nanut, M.; Kaur, K. NK cells shape pancreatic and oral tumor microenvironments; role in inhibition of tumor growth and metastasis. Semin. Cancer Biol. 2018, 53, 178–188. [Google Scholar] [CrossRef] [PubMed]
  16. Kaur, K.; Nanut, M.P.; Ko, M.W.; Safaie, T.; Kos, J.; Jewett, A. Natural killer cells target and differentiate cancer stem-like cells/undifferentiated tumors: Strategies to optimize their growth and expansion for effective cancer immunotherapy. Curr. Opin. Immunol. 2018, 51, 170–180. [Google Scholar] [CrossRef]
  17. Swanton, C. Intratumor heterogeneity: Evolution through space and time. Cancer Res. 2012, 72, 4875–4882. [Google Scholar] [CrossRef] [PubMed]
  18. Bashashati, A.; Ha, G.; Tone, A.; Ding, J.; Prentice, L.M.; Roth, A.; Rosner, J.; Shumansky, K.; Kalloger, S.; Senz, J.; et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 2013, 231, 21–34. [Google Scholar] [CrossRef] [PubMed]
  19. Castellarin, M.; Milne, K.; Zeng, T.; Tse, K.; Mayo, M.; Zhao, Y.; Webb, J.R.; Watson, P.H.; Nelson, B.H.; Holt, R.A. Clonal evolution of high-grade serous ovarian carcinoma from primary to recurrent disease. J. Pathol. 2013, 229, 515–524. [Google Scholar] [CrossRef] [PubMed]
  20. Schwarz, R.F.; Ng, C.K.; Cooke, S.L.; Newman, S.; Temple, J.; Piskorz, A.M.; Gale, D.; Sayal, K.; Murtaza, M.; Baldwin, P.J.; et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: A phylogenetic analysis. PLoS Med. 2015, 12, e1001789. [Google Scholar] [CrossRef] [PubMed]
  21. Lambrechts, S.; Smeets, D.; Moisse, M.; Braicu, E.I.; Vanderstichele, A.; Zhao, H.; Van Nieuwenhuysen, E.; Berns, E.; Sehouli, J.; Zeillinger, R.; et al. Genetic heterogeneity after first-line chemotherapy in high-grade serous ovarian cancer. Eur. J. Cancer 2016, 53, 51–64. [Google Scholar] [CrossRef] [PubMed]
  22. Latifi, A.; Abubaker, K.; Castrechini, N.; Ward, A.C.; Liongue, C.; Dobill, F.; Kumar, J.; Thompson, E.W.; Quinn, M.A.; Findlay, J.K.; et al. Cisplatin treatment of primary and metastatic epithelial ovarian carcinomas generates residual cells with mesenchymal stem cell-like profile. J. Cell. Biochem. 2011, 112, 2850–2864. [Google Scholar] [CrossRef] [PubMed]
  23. Thakur, B.; Ray, P. Cisplatin triggers cancer stem cell enrichment in platinum-resistant cells through NF-kappaB-TNFalpha-PIK3CA loop. J. Exp. Clin. Cancer Res. 2017, 36, 164. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, Y.; Cardenas, H.; Fang, F.; Condello, S.; Taverna, P.; Segar, M.; Liu, Y.; Nephew, K.P.; Matei, D. Epigenetic targeting of ovarian cancer stem cells. Cancer Res. 2014, 74, 4922–4936. [Google Scholar] [CrossRef] [PubMed]
  25. Steg, A.D.; Bevis, K.S.; Katre, A.A.; Ziebarth, A.; Dobbin, Z.C.; Alvarez, R.D.; Zhang, K.; Conner, M.; Landen, C.N. Stem cell pathways contribute to clinical chemoresistance in ovarian cancer. Clin. Cancer Res. 2012, 18, 869–881. [Google Scholar] [CrossRef] [PubMed]
  26. Yang, W.; Kim, D.; Kim, D.K.; Choi, K.U.; Suh, D.S.; Kim, J.H. Therapeutic Strategies for Targeting Ovarian Cancer Stem Cells. Int. J. Mol. Sci. 2021, 22, 5059. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, S.; Balch, C.; Chan, M.W.; Lai, H.C.; Matei, D.; Schilder, J.M.; Yan, P.S.; Huang, T.H.; Nephew, K.P. Identification and characterization of ovarian cancer-initiating cells from primary human tumors. Cancer Res. 2008, 68, 4311–4320. [Google Scholar] [CrossRef]
  28. Gao, M.Q.; Choi, Y.P.; Kang, S.; Youn, J.H.; Cho, N.H. CD24+ cells from hierarchically organized ovarian cancer are enriched in cancer stem cells. Oncogene 2010, 29, 2672–2680. [Google Scholar] [CrossRef] [PubMed]
  29. Baba, T.; Convery, P.A.; Matsumura, N.; Whitaker, R.S.; Kondoh, E.; Perry, T.; Huang, Z.; Bentley, R.C.; Mori, S.; Fujii, S.; et al. Epigenetic regulation of CD133 and tumorigenicity of CD133+ ovarian cancer cells. Oncogene 2009, 28, 209–218. [Google Scholar] [CrossRef] [PubMed]
  30. Silva, I.A.; Bai, S.; McLean, K.; Yang, K.; Griffith, K.; Thomas, D.; Ginestier, C.; Johnston, C.; Kueck, A.; Reynolds, R.K.; et al. Aldehyde dehydrogenase in combination with CD133 defines angiogenic ovarian cancer stem cells that portend poor patient survival. Cancer Res. 2011, 71, 3991–4001. [Google Scholar] [CrossRef] [PubMed]
  31. Robinson, M.; Gilbert, S.F.; Waters, J.A.; Lujano-Olazaba, O.; Lara, J.; Alexander, L.J.; Green, S.E.; Burkeen, G.A.; Patrus, O.; Sarwar, Z.; et al. Characterization of SOX2, OCT4 and NANOG in Ovarian Cancer Tumor-Initiating Cells. Cancers 2021, 13, 262. [Google Scholar] [CrossRef] [PubMed]
  32. Li, Y.R.; Ochoa, C.J.; Zhu, Y.; Kramer, A.; Wilson, M.; Fang, Y.; Chen, Y.; Singh, T.; Di Bernardo, G.; Zhu, E.; et al. Profiling ovarian cancer tumor and microenvironment during disease progression for cell-based immunotherapy design. iScience 2023, 26, 107952. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, L.; Zhang, F.; Cui, J.Y.; Chen, L.; Chen, Y.T.; Liu, B.W. CAFs enhance paclitaxel resistance by inducing EMT through the IL-6/JAK2/STAT3 pathway. Oncol. Rep. 2018, 39, 2081–2090. [Google Scholar] [CrossRef] [PubMed]
  34. Pasquier, J.; Gosset, M.; Geyl, C.; Hoarau-Véchot, J.; Chevrot, A.; Pocard, M.; Mirshahi, M.; Lis, R.; Rafii, A.; Touboul, C. CCL2/CCL5 secreted by the stroma induce IL-6/PYK2 dependent chemoresistance in ovarian cancer. Mol. Cancer 2018, 17, 47. [Google Scholar] [CrossRef]
  35. Wang, Y.; Niu, X.L.; Qu, Y.; Wu, J.; Zhu, Y.Q.; Sun, W.J.; Li, L.Z. Autocrine production of interleukin-6 confers cisplatin and paclitaxel resistance in ovarian cancer cells. Cancer Lett. 2010, 295, 110–123. [Google Scholar] [CrossRef] [PubMed]
  36. Leung, C.S.; Yeung, T.L.; Yip, K.P.; Wong, K.K.; Ho, S.Y.; Mangala, L.S.; Sood, A.K.; Lopez-Berestein, G.; Sheng, J.; Wong, S.T.; et al. Cancer-associated fibroblasts regulate endothelial adhesion protein LPP to promote ovarian cancer chemoresistance. J. Clin. Investig. 2018, 128, 589–606. [Google Scholar] [CrossRef] [PubMed]
  37. Zhang, D.; Ding, L.; Li, Y.; Ren, J.; Shi, G.; Wang, Y.; Zhao, S.; Ni, Y.; Hou, Y. Midkine derived from cancer-associated fibroblasts promotes cisplatin-resistance via up-regulation of the expression of lncRNA ANRIL in tumour cells. Sci. Rep. 2017, 7, 16231. [Google Scholar] [CrossRef] [PubMed]
  38. Gonzalez, V.D.; Samusik, N.; Chen, T.J.; Savig, E.S.; Aghaeepour, N.; Quigley, D.A.; Huang, Y.W.; Giangarrà, V.; Borowsky, A.D.; Hubbard, N.E.; et al. Commonly Occurring Cell Subsets in High-Grade Serous Ovarian Tumors Identified by Single-Cell Mass Cytometry. Cell Rep. 2018, 22, 1875–1888. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, Y.; Miao, C.; Jin, C.; Qiu, C.; Li, Y.; Sun, X.; Gao, M.; Lu, N.; Kong, B. SUSD2 promotes cancer metastasis and confers cisplatin resistance in high grade serous ovarian cancer. Exp. Cell Res. 2018, 363, 160–170. [Google Scholar] [CrossRef]
  40. Feng, T.; Wang, Y.; Lang, Y.; Zhang, Y. KDM5A promotes proliferation and EMT in ovarian cancer and closely correlates with PTX resistance. Mol. Med. Rep. 2017, 16, 3573–3580. [Google Scholar] [CrossRef]
  41. Sosulski, A.; Horn, H.; Zhang, L.; Coletti, C.; Vathipadiekal, V.; Castro, C.M.; Birrer, M.J.; Nagano, O.; Saya, H.; Lage, K.; et al. CD44 Splice Variant v8-10 as a Marker of Serous Ovarian Cancer Prognosis. PLoS ONE 2016, 11, e0156595. [Google Scholar] [CrossRef] [PubMed]
  42. Alhabbab, R.Y. Targeting Cancer Stem Cells by Genetically Engineered Chimeric Antigen Receptor T Cells. Front. Genet. 2020, 11, 312. [Google Scholar] [CrossRef] [PubMed]
  43. Wolf, N.K.; Kissiov, D.U.; Raulet, D.H. Roles of natural killer cells in immunity to cancer, and applications to immunotherapy. Nat. Rev. Immunol. 2023, 23, 90–105. [Google Scholar] [CrossRef] [PubMed]
  44. Chovatiya, N.; Kaur, K.; Huerta-Yepez, S.; Chen, P.C.; Neal, A.; DiBernardo, G.; Gumrukcu, S.; Memarzadeh, S.; Jewett, A. Inability of ovarian cancers to upregulate their MHC-class I surface expression marks their aggressiveness and increased susceptibility to NK cell-mediated cytotoxicity. Cancer Immunol. Immunother. 2022, 71, 2929–2941. [Google Scholar] [CrossRef]
  45. Coosemans, A.; Baert, T.; D’Heygere, V.; Wouters, R.; Laet, L.D.; Hoylandt, A.V.; Thirion, G.; Ceusters, J.; Laenen, A.; Vandecaveye, V.; et al. Increased Immunosuppression Is Related to Increased Amounts of Ascites and Inferior Prognosis in Ovarian Cancer. Anticancer Res. 2019, 39, 5953–5962. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, L.; Conejo-Garcia, J.R.; Katsaros, D.; Gimotty, P.A.; Massobrio, M.; Regnani, G.; Makrigiannakis, A.; Gray, H.; Schlienger, K.; Liebman, M.N.; et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 2003, 348, 203–213. [Google Scholar] [CrossRef] [PubMed]
  47. Rabinowich, H.; Suminami, Y.; Reichert, T.E.; Crowley-Nowick, P.; Bell, M.; Edwards, R.; Whiteside, T.L. Expression of cytokine genes or proteins and signaling molecules in lymphocytes associated with human ovarian carcinoma. Int. J. Cancer 1996, 68, 276–284. [Google Scholar] [CrossRef]
  48. Ioannides, C.G.; Platsoucas, C.D.; Rashed, S.; Wharton, J.T.; Edwards, C.L.; Freedman, R.S. Tumor cytolysis by lymphocytes infiltrating ovarian malignant ascites. Cancer Res. 1991, 51, 4257–4265. [Google Scholar] [PubMed]
  49. Lukesova, S.; Vroblova, V.; Tosner, J.; Kopecky, J.; Sedlakova, I.; Čermáková, E.; Vokurkova, D.; Kopecky, O. Comparative study of various subpopulations of cytotoxic cells in blood and ascites from patients with ovarian carcinoma. Contemp. Oncol. 2015, 19, 290–299. [Google Scholar] [CrossRef] [PubMed]
  50. Lai, P.; Rabinowich, H.; Crowley-Nowick, P.A.; Bell, M.C.; Mantovani, G.; Whiteside, T.L. Alterations in expression and function of signal-transducing proteins in tumor-associated T and natural killer cells in patients with ovarian carcinoma. Clin. Cancer Res. 1996, 2, 161–173. [Google Scholar] [PubMed]
  51. Introna, M.; Allavena, P.; Biondi, A.; Colombo, N.; Villa, A.; Mantovani, A. Defective natural killer activity within human ovarian tumors: Low numbers of morphologically defined effectors present in situ. J. Natl. Cancer Inst. 1983, 70, 21–26. [Google Scholar] [PubMed]
  52. Pesce, S.; Tabellini, G.; Cantoni, C.; Patrizi, O.; Coltrini, D.; Rampinelli, F.; Matta, J.; Vivier, E.; Moretta, A.; Parolini, S.; et al. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology 2015, 4, e1001224. [Google Scholar] [CrossRef] [PubMed]
  53. Yunusova, N.V.; Stakheyeva, M.N.; Molchanov, S.V.; Afanas’ev, S.G.; Tsydenova, A.A.; Kolomiets, L.A.; Cherdyntseva, N.V. Functional activity of natural killer cells in biological fluids in patients with colorectal and ovarian cancers. Cent. Eur. J. Immunol. 2018, 43, 26–32. [Google Scholar] [CrossRef] [PubMed]
  54. Berek, J.S.; Bast, R.C., Jr.; Lichtenstein, A.; Hacker, N.F.; Spina, C.A.; Lagasse, L.D.; Knapp, R.C.; Zighelboim, J. Lymphocyte cytotoxicity in the peritoneal cavity and blood of patients with ovarian cancer. Obs. Gynecol. 1984, 64, 708–714. [Google Scholar]
  55. Buckanovich, R.J.; Facciabene, A.; Kim, S.; Benencia, F.; Sasaroli, D.; Balint, K.; Katsaros, D.; O’Brien-Jenkins, A.; Gimotty, P.A.; Coukos, G. Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat. Med. 2008, 14, 28–36. [Google Scholar] [CrossRef] [PubMed]
  56. Webb, J.R.; Milne, K.; Watson, P.; Deleeuw, R.J.; Nelson, B.H. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer. Clin. Cancer Res. 2014, 20, 434–444. [Google Scholar] [CrossRef] [PubMed]
  57. Hoogstad-van Evert, J.S.; Maas, R.J.; van der Meer, J.; Cany, J.; van der Steen, S.; Jansen, J.H.; Miller, J.S.; Bekkers, R.; Hobo, W.; Massuger, L.; et al. Peritoneal NK cells are responsive to IL-15 and percentages are correlated with outcome in advanced ovarian cancer patients. Oncotarget 2018, 9, 34810–34820. [Google Scholar] [CrossRef] [PubMed]
  58. Felices, M.; Chu, S.; Kodal, B.; Bendzick, L.; Ryan, C.; Lenvik, A.J.; Boylan, K.L.M.; Wong, H.C.; Skubitz, A.P.N.; Miller, J.S.; et al. IL-15 super-agonist (ALT-803) enhances natural killer (NK) cell function against ovarian cancer. Gynecol. Oncol. 2017, 145, 453–461. [Google Scholar] [CrossRef] [PubMed]
  59. Wei, S.; Kryczek, I.; Zou, L.; Daniel, B.; Cheng, P.; Mottram, P.; Curiel, T.; Lange, A.; Zou, W. Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma. Cancer Res. 2005, 65, 5020–5026. [Google Scholar] [CrossRef] [PubMed]
  60. Barber, A.; Rynda, A.; Sentman, C.L. Chimeric NKG2D expressing T cells eliminate immunosuppression and activate immunity within the ovarian tumor microenvironment. J. Immunol. 2009, 183, 6939–6947. [Google Scholar] [CrossRef] [PubMed]
  61. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef]
  62. Sato, E.; Olson, S.H.; Ahn, J.; Bundy, B.; Nishikawa, H.; Qian, F.; Jungbluth, A.A.; Frosina, D.; Gnjatic, S.; Ambrosone, C.; et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc. Natl. Acad. Sci. USA 2005, 102, 18538–18543. [Google Scholar] [CrossRef] [PubMed]
  63. Pujade-Lauraine, E.; Hilpert, F.; Weber, B.; Reuss, A.; Poveda, A.; Kristensen, G.; Sorio, R.; Vergote, I.; Witteveen, P.; Bamias, A.; et al. Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. J. Clin. Oncol. 2014, 32, 1302–1308. [Google Scholar] [CrossRef]
  64. Aghajanian, C.; Blank, S.V.; Goff, B.A.; Judson, P.L.; Teneriello, M.G.; Husain, A.; Sovak, M.A.; Yi, J.; Nycum, L.R. OCEANS: A randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J. Clin. Oncol. 2012, 30, 2039–2045. [Google Scholar] [CrossRef]
  65. Oza, A.M.; Cook, A.D.; Pfisterer, J.; Embleton, A.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; et al. Standard chemotherapy with or without bevacizumab for women with newly diagnosed ovarian cancer (ICON7): Overall survival results of a phase 3 randomised trial. Lancet Oncol. 2015, 16, 928–936. [Google Scholar] [CrossRef] [PubMed]
  66. Hamanishi, J.; Mandai, M.; Ikeda, T.; Minami, M.; Kawaguchi, A.; Murayama, T.; Kanai, M.; Mori, Y.; Matsumoto, S.; Chikuma, S.; et al. Safety and Antitumor Activity of Anti-PD-1 Antibody, Nivolumab, in Patients With Platinum-Resistant Ovarian Cancer. J. Clin. Oncol. 2015, 33, 4015–4022. [Google Scholar] [CrossRef] [PubMed]
  67. Matulonis, U.A.; Shapira-Frommer, R.; Santin, A.D.; Lisyanskaya, A.S.; Pignata, S.; Vergote, I.; Raspagliesi, F.; Sonke, G.S.; Birrer, M.; Provencher, D.M.; et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: Results from the phase II KEYNOTE-100 study. Ann. Oncol. 2019, 30, 1080–1087. [Google Scholar] [CrossRef] [PubMed]
  68. Zamarin, D.; Burger, R.A.; Sill, M.W.; Powell, D.J., Jr.; Lankes, H.A.; Feldman, M.D.; Zivanovic, O.; Gunderson, C.; Ko, E.; Mathews, C.; et al. Randomized Phase II Trial of Nivolumab Versus Nivolumab and Ipilimumab for Recurrent or Persistent Ovarian Cancer: An NRG Oncology Study. J. Clin. Oncol. 2020, 38, 1814–1823. [Google Scholar] [CrossRef] [PubMed]
  69. Leffers, N.; Lambeck, A.J.; Gooden, M.J.; Hoogeboom, B.N.; Wolf, R.; Hamming, I.E.; Hepkema, B.G.; Willemse, P.H.; Molmans, B.H.; Hollema, H.; et al. Immunization with a P53 synthetic long peptide vaccine induces P53-specific immune responses in ovarian cancer patients, a phase II trial. Int. J. Cancer 2009, 125, 2104–2113. [Google Scholar] [CrossRef]
  70. Vermeij, R.; Leffers, N.; Hoogeboom, B.N.; Hamming, I.L.; Wolf, R.; Reyners, A.K.; Molmans, B.H.; Hollema, H.; Bart, J.; Drijfhout, J.W.; et al. Potentiation of a p53-SLP vaccine by cyclophosphamide in ovarian cancer: A single-arm phase II study. Int. J. Cancer 2012, 131, E670–E680. [Google Scholar] [CrossRef] [PubMed]
  71. Schoutrop, E.; El-Serafi, I.; Poiret, T.; Zhao, Y.; Gultekin, O.; He, R.; Moyano-Galceran, L.; Carlson, J.W.; Lehti, K.; Hassan, M.; et al. Mesothelin-Specific CAR T Cells Target Ovarian Cancer. Cancer Res. 2021, 81, 3022–3035. [Google Scholar] [CrossRef] [PubMed]
  72. Geller, M.A.; Knorr, D.A.; Hermanson, D.A.; Pribyl, L.; Bendzick, L.; McCullar, V.; Miller, J.S.; Kaufman, D.S. Intraperitoneal delivery of human natural killer cells for treatment of ovarian cancer in a mouse xenograft model. Cytotherapy 2013, 15, 1297–1306. [Google Scholar] [CrossRef] [PubMed]
  73. Hermanson, D.L.; Bendzick, L.; Pribyl, L.; McCullar, V.; Vogel, R.I.; Miller, J.S.; Geller, M.A.; Kaufman, D.S. Induced Pluripotent Stem Cell-Derived Natural Killer Cells for Treatment of Ovarian Cancer. Stem Cells 2016, 34, 93–101. [Google Scholar] [CrossRef] [PubMed]
  74. Uppendahl, L.D.; Dahl, C.M.; Miller, J.S.; Felices, M.; Geller, M.A. Natural Killer Cell-Based Immunotherapy in Gynecologic Malignancy: A Review. Front. Immunol. 2017, 8, 1825. [Google Scholar] [CrossRef] [PubMed]
  75. Hoogstad-van Evert, J.S.; Cany, J.; van den Brand, D.; Oudenampsen, M.; Brock, R.; Torensma, R.; Bekkers, R.L.; Jansen, J.H.; Massuger, L.F.; Dolstra, H. Umbilical cord blood CD34(+) progenitor-derived NK cells efficiently kill ovarian cancer spheroids and intraperitoneal tumors in NOD/SCID/IL2Rg(null) mice. Oncoimmunology 2017, 6, e1320630. [Google Scholar] [CrossRef] [PubMed]
  76. Nham, T.; Poznanski, S.M.; Fan, I.Y.; Shenouda, M.M.; Chew, M.V.; Lee, A.J.; Vahedi, F.; Karimi, Y.; Butcher, M.; Lee, D.A.; et al. Ex vivo-expanded NK cells from blood and ascites of ovarian cancer patients are cytotoxic against autologous primary ovarian cancer cells. Cancer Immunol. Immunother. 2018, 67, 575–587. [Google Scholar] [CrossRef] [PubMed]
  77. Sun, Y.; Yao, Z.; Zhao, Z.; Xiao, H.; Xia, M.; Zhu, X.; Jiang, X.; Sun, C. Natural killer cells inhibit metastasis of ovarian carcinoma cells and show therapeutic effects in a murine model of ovarian cancer. Exp. Ther. Med. 2018, 16, 1071–1078. [Google Scholar] [CrossRef] [PubMed]
  78. Xie, S.; Chen, J.; Zhang, M.; Wu, Z. Allogenic natural killer cell immunotherapy of sizeable ovarian cancer: A case report. Mol. Clin. Oncol. 2017, 6, 903–906. [Google Scholar] [CrossRef] [PubMed]
  79. Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef]
  80. Tseng, H.C.; Arasteh, A.; Kaur, K.; Kozlowska, A.; Topchyan, P.; Jewett, A. Differential Cytotoxicity but Augmented IFN-gamma Secretion by NK Cells after Interaction with Monocytes from Humans, and Those from Wild Type and Myeloid-Specific COX-2 Knockout Mice. Front. Immunol. 2015, 6, 259. [Google Scholar] [CrossRef] [PubMed]
  81. Cacalano, N.A.; Le, D.; Paranjpe, A.; Wang, M.Y.; Fernandez, A.; Evazyan, T.; Park, N.H.; Jewett, A. Regulation of IGFBP6 gene and protein is mediated by the inverse expression and function of c-jun N-terminal kinase (JNK) and NFkappaB in a model of oral tumor cells. Apoptosis 2008, 13, 1439–1449. [Google Scholar] [CrossRef] [PubMed]
  82. Jewett, A.; Cacalano, N.A.; Teruel, A.; Romero, M.; Rashedi, M.; Wang, M.; Nakamura, H. Inhibition of nuclear factor kappa B (NFkappaB) activity in oral tumor cells prevents depletion of NK cells and increases their functional activation. Cancer Immunol. Immunother. 2006, 55, 1052–1063. [Google Scholar] [CrossRef]
  83. Jewett, A.; Wang, M.Y.; Teruel, A.; Poupak, Z.; Bostanian, Z.; Park, N.H. Cytokine dependent inverse regulation of CD54 (ICAM1) and major histocompatibility complex class I antigens by nuclear factor kappaB in HEp2 tumor cell line: Effect on the function of natural killer cells. Hum. Immunol. 2003, 64, 505–520. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, J.; Cherwinski, H.; Spies, T.; Phillips, J.H.; Lanier, L.L. DAP10 and DAP12 form distinct, but functionally cooperative, receptor complexes in natural killer cells. J. Exp. Med. 2000, 192, 1059–1068. [Google Scholar] [CrossRef] [PubMed]
  85. Bui, V.T.; Tseng, H.C.; Kozlowska, A.; Maung, P.O.; Kaur, K.; Topchyan, P.; Jewett, A. Augmented IFN-γ and TNF-α Induced by Probiotic Bacteria in NK Cells Mediate Differentiation of Stem-Like Tumors Leading to Inhibition of Tumor Growth and Reduction in Inflammatory Cytokine Release; Regulation by IL-10. Front. Immunol. 2015, 6, 576. [Google Scholar] [CrossRef] [PubMed]
  86. Kaur, K.; Kozlowska, A.K.; Topchyan, P.; Ko, M.W.; Ohanian, N.; Chiang, J.; Cook, J.; Maung, P.O.; Park, S.H.; Cacalano, N.; et al. Probiotic-Treated Super-Charged NK Cells Efficiently Clear Poorly Differentiated Pancreatic Tumors in Hu-BLT Mice. Cancers 2019, 12, 63. [Google Scholar] [CrossRef] [PubMed]
  87. Hamilton, T.C.; Young, R.C.; McKoy, W.M.; Grotzinger, K.R.; Green, J.A.; Chu, E.W.; Whang-Peng, J.; Rogan, A.M.; Green, W.R.; Ozols, R.F. Characterization of a human ovarian carcinoma cell line (NIH:OVCAR-3) with androgen and estrogen receptors. Cancer Res. 1983, 43, 5379–5389. [Google Scholar] [PubMed]
  88. Patankar, M.S.; Jing, Y.; Morrison, J.C.; Belisle, J.A.; Lattanzio, F.A.; Deng, Y.; Wong, N.K.; Morris, H.R.; Dell, A.; Clark, G.F. Potent suppression of natural killer cell response mediated by the ovarian tumor marker CA125. Gynecol. Oncol. 2005, 99, 704–713. [Google Scholar] [CrossRef] [PubMed]
  89. Gubbels, J.A.; Felder, M.; Horibata, S.; Belisle, J.A.; Kapur, A.; Holden, H.; Petrie, S.; Migneault, M.; Rancourt, C.; Connor, J.P.; et al. MUC16 provides immune protection by inhibiting synapse formation between NK and ovarian tumor cells. Mol. Cancer 2010, 9, 11. [Google Scholar] [CrossRef]
  90. Gubbels, J.A.; Belisle, J.; Onda, M.; Rancourt, C.; Migneault, M.; Ho, M.; Bera, T.K.; Connor, J.; Sathyanarayana, B.K.; Lee, B.; et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol. Cancer 2006, 5, 50. [Google Scholar] [CrossRef] [PubMed]
  91. Johnson, S.W.; Laub, P.B.; Beesley, J.S.; Ozols, R.F.; Hamilton, T.C. Increased platinum-DNA damage tolerance is associated with cisplatin resistance and cross-resistance to various chemotherapeutic agents in unrelated human ovarian cancer cell lines. Cancer Res. 1997, 57, 850–856. [Google Scholar] [PubMed]
  92. Mitra, A.K.; Davis, D.A.; Tomar, S.; Roy, L.; Gurler, H.; Xie, J.; Lantvit, D.D.; Cardenas, H.; Fang, F.; Liu, Y.; et al. In vivo tumor growth of high-grade serous ovarian cancer cell lines. Gynecol. Oncol. 2015, 138, 372–377. [Google Scholar] [CrossRef] [PubMed]
  93. Gottschalk, N.; Lang, S.; Kimmig, R.; Singh, M.; Brandau, S. Monocytes and the 38kDa-antigen of mycobacterium tuberculosis modulate natural killer cell activity and their cytolysis directed against ovarian cancer cell lines. BMC Cancer 2012, 12, 451. [Google Scholar] [CrossRef] [PubMed]
  94. Haley, J.; Tomar, S.; Pulliam, N.; Xiong, S.; Perkins, S.M.; Karpf, A.R.; Mitra, S.; Nephew, K.P.; Mitra, A.K. Functional characterization of a panel of high-grade serous ovarian cancer cell lines as representative experimental models of the disease. Oncotarget 2016, 7, 32810–32820. [Google Scholar] [CrossRef] [PubMed]
  95. Gao, Y.; Foster, R.; Yang, X.; Feng, Y.; Shen, J.K.; Mankin, H.J.; Hornicek, F.J.; Amiji, M.M.; Duan, Z. Up-regulation of CD44 in the development of metastasis, recurrence and drug resistance of ovarian cancer. Oncotarget 2015, 6, 9313–9326. [Google Scholar] [CrossRef] [PubMed]
  96. Natoli, M.; Bonito, N.; Robinson, J.D.; Ghaem-Maghami, S.; Mao, Y. Human ovarian cancer intrinsic mechanisms regulate lymphocyte activation in response to immune checkpoint blockade. Cancer Immunol. Immunother. 2020, 69, 1391–1401. [Google Scholar] [CrossRef] [PubMed]
  97. Fogh, J.; Fogh, J.M.; Orfeo, T. One hundred and twenty-seven cultured human tumor cell lines producing tumors in nude mice. J. Natl. Cancer Inst. 1977, 59, 221–226. [Google Scholar] [CrossRef] [PubMed]
  98. Hoogstad-van Evert, J.S.; Bekkers, R.; Ottevanger, N.; Jansen, J.H.; Massuger, L.; Dolstra, H. Harnessing natural killer cells for the treatment of ovarian cancer. Gynecol. Oncol. 2020, 157, 810–816. [Google Scholar] [CrossRef]
  99. Gonzalez, V.D.; Huang, Y.-W.; Chen, S.-Y.; Delgado-Gonzalez, A.; Donoso, K.; Gentles, A.; Sachs, K.; Porpiglia, E.; Fantl, W.J. High-Grade Serous Ovarian Tumor Cells Modulate NK Cell Function to Create an Immune-Tolerant Microenvironment. bioRxiv, 2020; preprint. [Google Scholar] [CrossRef]
  100. Beaufort, C.M.; Helmijr, J.C.; Piskorz, A.M.; Hoogstraat, M.; Ruigrok-Ritstier, K.; Besselink, N.; Murtaza, M.; van IJcken, W.F.; Heine, A.A.; Smid, M.; et al. Ovarian cancer cell line panel (OCCP): Clinical importance of in vitro morphological subtypes. PLoS ONE 2014, 9, e103988. [Google Scholar] [CrossRef] [PubMed]
  101. Cannistra, S.A.; DeFranzo, B.; Niloff, J.; Ottensmeir, C. Functional heterogeneity of CD44 molecules in ovarian cancer cell lines. Clin. Cancer Res. 1995, 1, 333–342. [Google Scholar] [PubMed]
  102. Cannistra, S.A.; Kansas, G.S.; Niloff, J.; DeFranzo, B.; Kim, Y.; Ottensmeier, C. Binding of ovarian cancer cells to peritoneal mesothelium in vitro is partly mediated by CD44H. Cancer Res. 1993, 53, 3830–3838. [Google Scholar] [PubMed]
  103. Jewett, A.; Kos, J.; Kaur, K.; Turnsek, T.L.; Breznik, B.; Senjor, E.; Wong, P.; Nguyen, K.Y.; Ko, M.W. Multiple Defects of Natural Killer Cells in Cancer Patients: Anarchy, Dysregulated Systemic Immunity, and Immunosuppression in Metastatic Cancer. Crit. Rev. Immunol. 2020, 40, 93–133. [Google Scholar] [CrossRef] [PubMed]
  104. Tseng, H.C.; Arasteh, A.; Paranjpe, A.; Teruel, A.; Yang, W.; Behel, A.; Alva, J.A.; Walter, G.; Head, C.; Ishikawa, T.O.; et al. Increased lysis of stem cells but not their differentiated cells by natural killer cells; de-differentiation or reprogramming activates NK cells. PLoS ONE 2010, 5, e11590. [Google Scholar] [CrossRef] [PubMed]
  105. Griesinger, L.; Nyarko-Odoom, A.; Martinez, S.A.; Shen, N.W.; Ring, K.L.; Gaughan, E.M.; Mills, A.M. PD-L1 and MHC Class I Expression in High-grade Ovarian Cancers, Including Platinum-resistant Recurrences Treated With Checkpoint Inhibitor Therapy. Appl. Immunohistochem. Mol. Morphol. 2023, 31, 197–203. [Google Scholar] [CrossRef] [PubMed]
  106. Schuster, H.; Peper, J.K.; Bösmüller, H.C.; Röhle, K.; Backert, L.; Bilich, T.; Ney, B.; Löffler, M.W.; Kowalewski, D.J.; Trautwein, N.; et al. The immunopeptidomic landscape of ovarian carcinomas. Proc. Natl. Acad. Sci. USA 2017, 114, E9942–E9951. [Google Scholar] [CrossRef]
  107. Aust, S.; Felix, S.; Auer, K.; Bachmayr-Heyda, A.; Kenner, L.; Dekan, S.; Meier, S.M.; Gerner, C.; Grimm, C.; Pils, D. Absence of PD-L1 on tumor cells is associated with reduced MHC I expression and PD-L1 expression increases in recurrent serous ovarian cancer. Sci. Rep. 2017, 7, 42929. [Google Scholar] [CrossRef] [PubMed]
  108. Turner, T.B.; Meza-Perez, S.; Londoño, A.; Katre, A.; Peabody, J.E.; Smith, H.J.; Forero, A.; Norian, L.A.; Straughn, J.M., Jr.; Buchsbaum, D.J.; et al. Epigenetic modifiers upregulate MHC II and impede ovarian cancer tumor growth. Oncotarget 2017, 8, 44159–44170. [Google Scholar] [CrossRef] [PubMed]
  109. Lin, S.Y.; Hang, J.F.; Lai, C.R.; Chan, I.S.; Shih, Y.C.; Jiang, L.Y.; Chang, Y.H.; Chen, Y.J. Loss of Major Histocompatibility Complex Class I, CD8 + Tumor-infiltrating Lymphocytes, and PD-L1 Expression in Ovarian Clear Cell Carcinoma. Am. J. Surg. Pathol. 2023, 47, 124–130. [Google Scholar] [CrossRef] [PubMed]
  110. Cornel, A.M.; Mimpen, I.L.; Nierkens, S. MHC Class I Downregulation in Cancer: Underlying Mechanisms and Potential Targets for Cancer Immunotherapy. Cancers 2020, 12, 1760. [Google Scholar] [CrossRef] [PubMed]
  111. Callahan, M.J.; Nagymanyoki, Z.; Bonome, T.; Johnson, M.E.; Litkouhi, B.; Sullivan, E.H.; Hirsch, M.S.; Matulonis, U.A.; Liu, J.; Birrer, M.J.; et al. Increased HLA-DMB expression in the tumor epithelium is associated with increased CTL infiltration and improved prognosis in advanced-stage serous ovarian cancer. Clin. Cancer Res. 2008, 14, 7667–7673. [Google Scholar] [CrossRef] [PubMed]
  112. Brunekreeft, K.L.; Paijens, S.T.; Wouters, M.C.A.; Komdeur, F.L.; Eggink, F.A.; Lubbers, J.M.; Workel, H.H.; Van Der Slikke, E.C.; Pröpper, N.E.J.; Leffers, N.; et al. Deep immune profiling of ovarian tumors identifies minimal MHC-I expression after neoadjuvant chemotherapy as negatively associated with T-cell-dependent outcome. Oncoimmunology 2020, 9, 1760705. [Google Scholar] [CrossRef] [PubMed]
  113. Palmer, J.M.; Rajasekaran, K.; Thakar, M.S.; Malarkannan, S. Clinical relevance of natural killer cells following hematopoietic stem cell transplantation. J. Cancer 2013, 4, 25–35. [Google Scholar] [CrossRef] [PubMed]
  114. Fildes, J.E.; Yonan, N.; Leonard, C.T. Natural killer cells and lung transplantation, roles in rejection, infection, and tolerance. Transpl. Immunol. 2008, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  115. Farag, S.S.; Caligiuri, M.A. Human natural killer cell development and biology. Blood Rev. 2006, 20, 123–137. [Google Scholar] [CrossRef] [PubMed]
  116. Burke, S.; Lakshmikanth, T.; Colucci, F.; Carbone, E. New views on natural killer cell-based immunotherapy for melanoma treatment. Trends Immunol. 2010, 31, 339–345. [Google Scholar] [CrossRef] [PubMed]
  117. Jurisic, V.; Srdic, T.; Konjevic, G.; Markovic, O.; Colovic, M. Clinical stage-depending decrease of NK cell activity in multiple myeloma patients. Med. Oncol. 2007, 24, 312–317. [Google Scholar] [CrossRef] [PubMed]
  118. Larsen, S.K.; Gao, Y.; Basse, P.H. NK cells in the tumor microenvironment. Crit. Rev. Oncog. 2014, 19, 91–105. [Google Scholar] [CrossRef] [PubMed]
  119. 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, 1795–1799. [Google Scholar] [CrossRef] [PubMed]
  120. Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [PubMed]
  121. Venstrom, J.M.; Pittari, G.; Gooley, T.A.; Chewning, J.H.; Spellman, S.; Haagenson, M.; Gallagher, M.M.; Malkki, M.; Petersdorf, E.; Dupont, B.; et al. HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N. Engl. J. Med. 2012, 367, 805–816. [Google Scholar] [CrossRef] [PubMed]
  122. Iliopoulou, E.G.; Kountourakis, P.; Karamouzis, M.V.; Doufexis, D.; Ardavanis, A.; Baxevanis, C.N.; Rigatos, G.; Papamichail, M.; Perez, S.A. A phase I trial of adoptive transfer of allogeneic natural killer cells in patients with advanced non-small cell lung cancer. Cancer Immunol. Immunother. 2010, 59, 1781–1789. [Google Scholar] [CrossRef] [PubMed]
  123. Miller, J.S.; Soignier, Y.; Panoskaltsis-Mortari, A.; McNearney, S.A.; Yun, G.H.; Fautsch, S.K.; McKenna, D.; Le, C.; Defor, T.E.; Burns, L.J.; et al. Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005, 105, 3051–3057. [Google Scholar] [CrossRef] [PubMed]
  124. Re, F.; Staudacher, C.; Zamai, L.; Vecchio, V.; Bregni, M. Killer cell Ig-like receptors ligand-mismatched, alloreactive natural killer cells lyse primary solid tumors. Cancer 2006, 107, 640–648. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, H.; Blum, R.H.; Bjordahl, R.; Gaidarova, S.; Rogers, P.; Lee, T.T.; Abujarour, R.; Bonello, G.B.; Wu, J.; Tsai, P.F.; et al. Pluripotent stem cell-derived NK cells with high-affinity noncleavable CD16a mediate improved antitumor activity. Blood 2020, 135, 399–410. [Google Scholar] [CrossRef] [PubMed]
  126. Chu, J.; Gao, F.; Yan, M.; Zhao, S.; Yan, Z.; Shi, B.; Liu, Y. Natural killer cells: A promising immunotherapy for cancer. J. Transl. Med. 2022, 20, 240. [Google Scholar] [CrossRef] [PubMed]
  127. Portale, F.; Di Mitri, D. NK Cells in Cancer: Mechanisms of Dysfunction and Therapeutic Potential. Int. J. Mol. Sci. 2023, 24, 9521. [Google Scholar] [CrossRef] [PubMed]
  128. Kaur, K.; Cook, J.; Park, S.H.; Topchyan, P.; Kozlowska, A.; Ohanian, N.; Fang, C.; Nishimura, I.; Jewett, A. Novel Strategy to Expand Super-Charged NK Cells with Significant Potential to Lyse and Differentiate Cancer Stem Cells: Differences in NK Expansion and Function between Healthy and Cancer Patients. Front. Immunol. 2017, 8, 297. [Google Scholar] [CrossRef] [PubMed]
  129. Kaur, K.; Ko, M.W.; Ohanian, N.; Cook, J.; Jewett, A. Osteoclast-expanded super-charged NK-cells preferentially select and expand CD8+ T cells. Sci. Rep. 2020, 10, 20363. [Google Scholar] [CrossRef]
  130. Kaur, K.; Chen, P.-C.; Ko, M.-W.; Mei, A.; Senjor, E.; Malarkannan, S.; Kos, J.; Jewett, A. Sequential therapy with supercharged NK cells with either chemotherapy drug cisplatin or anti-PD-1 antibody decreases the tumor size and significantly enhances the NK function in Hu-BLT mice. Front. Immunol. 2023, 14, 1132807. [Google Scholar] [CrossRef]
  131. Kaur, K.; Ko, M.-W.; Chen, F.; Jewett, A. Defective NK cell expansion, cytotoxicity, and lack of ability to differentiate tumors from a pancreatic cancer patient in a long term follow-up: Implication in the progression of cancer. Cancer Immunol. Immunother. 2022, 71, 1033–1047. [Google Scholar] [CrossRef] [PubMed]
  132. Kaur, K.; Safaie, T.; Ko, M.-W.; Wang, Y.; Jewett, A. ADCC against MICA/B Is Mediated against Differentiated Oral and Pancreatic and Not Stem-Like/Poorly Differentiated Tumors by the NK Cells; Loss in Cancer Patients due to Down-Modulation of CD16 Receptor. Cancers 2021, 13, 239. [Google Scholar] [CrossRef] [PubMed]
  133. Kaur, K.; Ko, M.-W.; Chen, P.-C.; Breznik, B.; Senjor, E.; Wong, P.; Wang, Y.; Chovatiya, N.; Jewett, A. Probiotics in Health and Disease: Distinct Roles of Different Strains in Natural Killer Cell Activation and Regulation. Crit. Rev. Immunol. 2021, 41, 1–19. [Google Scholar] [CrossRef]
  134. Jewett, A.; Kos, J.; Kaur, K.; Safaei, T.; Sutanto, C.; Chen, W.; Wong, P.; Namagerdi, A.K.; Fang, C.; Fong, Y.; et al. Natural Killer Cells: Diverse Functions in Tumor Immunity and Defects in Pre-neoplastic and Neoplastic Stages of Tumorigenesis. Mol. Ther. Oncolytics 2020, 16, 41–52. [Google Scholar] [CrossRef] [PubMed]
  135. Kaur, K.; Topchyan, P.; Kozlowska, A.K.; Ohanian, N.; Chiang, J.; Maung, P.O.; Park, S.-H.; Ko, M.-W.; Fang, C.; Nishimura, I.; et al. Super-charged NK cells inhibit growth and progression of stem-like/poorly differentiated oral tumors in vivo in humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs. OncoImmunology 2018, 7, e1426518. [Google Scholar] [CrossRef] [PubMed]
  136. Kaur, K.; Chang, H.-H.; Cook, J.; Eibl, G.; Jewett, A. Suppression of Gingival NK Cells in Precancerous and Cancerous Stages of Pancreatic Cancer in KC and BLT-Humanized Mice. Front. Immunol. 2017, 8, 1606. [Google Scholar] [CrossRef]
  137. Tseng, H.C.; Kanayama, K.; Kaur, K.; Park, S.H.; Park, S.; Kozlowska, A.; Sun, S.; McKenna, C.E.; Nishimura, I.; Jewett, A. Bisphosphonate-induced differential modulation of immune cell function in gingiva and bone marrow in vivo: Role in osteoclast-mediated NK cell activation. Oncotarget 2015, 6, 20002–20025. [Google Scholar] [CrossRef]
  138. Li, H.; Hong, S.; Qian, J.; Zheng, Y.; Yang, J.; Yi, Q. Cross talk between the bone and immune systems: Osteoclasts function as antigen-presenting cells and activate CD4+ and CD8+ T cells. Blood 2010, 116, 210–217. [Google Scholar] [CrossRef] [PubMed]
  139. Dong, H.; Rowland, I.; Yaqoob, P. Comparative effects of six probiotic strains on immune function in vitro. Br. J. Nutr. 2012, 108, 459–470. [Google Scholar] [CrossRef]
  140. Chiang, J.; Chen, P.C.; Pham, J.; Nguyen, C.Q.; Kaur, K.; Raman, S.S.; Jewett, A. Characterizing hepatocellular carcinoma stem markers and their corresponding susceptibility to NK-cell based immunotherapy. Front. Immunol. 2023, 14, 1284669. [Google Scholar] [CrossRef]
  141. Liu, E.; Marin, D.; Banerjee, P.; Macapinlac, H.A.; Thompson, P.; Basar, R.; Nassif Kerbauy, L.; Overman, B.; Thall, P.; Kaplan, M.; et al. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N. Engl. J. Med. 2020, 382, 545–553. [Google Scholar] [CrossRef] [PubMed]
  142. Terranova-Barberio, M.; Pawlowska, N.; Dhawan, M.; Moasser, M.; Chien, A.J.; Melisko, M.E.; Rugo, H.; Rahimi, R.; Deal, T.; Daud, A.; et al. Exhausted T cell signature predicts immunotherapy response in ER-positive breast cancer. Nat. Commun. 2020, 11, 3584. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, E.X.; Jonker, D.J.; Loree, J.M.; Kennecke, H.F.; Berry, S.R.; Couture, F.; Ahmad, C.E.; Goffin, J.R.; Kavan, P.; Harb, M.; et al. Effect of Combined Immune Checkpoint Inhibition vs Best Supportive Care Alone in Patients With Advanced Colorectal Cancer: The Canadian Cancer Trials Group CO.26 Study. JAMA Oncol. 2020, 6, 831–838. [Google Scholar] [CrossRef] [PubMed]
  144. Rosenberg, S.A.; Restifo, N.P. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015, 348, 62–68. [Google Scholar] [CrossRef] [PubMed]
  145. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef]
  146. Liem, N.T.; Van Phong, N.; Kien, N.T.; Anh, B.V.; Huyen, T.L.; Thao, C.T.; Tu, N.D.; Hiep, D.T.; Hoai Thu, D.T.; Nhung, H.T.M. Phase I Clinical Trial Using Autologous Ex Vivo Expanded NK Cells and Cytotoxic T Lymphocytes for Cancer Treatment in Vietnam. Int. J. Mol. Sci. 2019, 20, 3166. [Google Scholar] [CrossRef] [PubMed]
  147. Leivas, A.; Perez-Martinez, A.; Blanchard, M.J.; Martín-Clavero, E.; Fernández, L.; Lahuerta, J.J.; Martinez-Lopez, J. Novel treatment strategy with autologous activated and expanded natural killer cells plus anti-myeloma drugs for multiple myeloma. Oncoimmunology 2016, 5, e1250051. [Google Scholar] [CrossRef] [PubMed]
  148. Lee, J.H.; Lee, J.H.; Lim, Y.S.; Yeon, J.E.; Song, T.J.; Yu, S.J.; Gwak, G.Y.; Kim, K.M.; Kim, Y.J.; Lee, J.W.; et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology 2015, 148, 1383–1391.e1386. [Google Scholar] [CrossRef] [PubMed]
  149. Sakamoto, N.; Ishikawa, T.; Kokura, S.; Okayama, T.; Oka, K.; Ideno, M.; Sakai, F.; Kato, A.; Tanabe, M.; Enoki, T.; et al. Phase I clinical trial of autologous NK cell therapy using novel expansion method in patients with advanced digestive cancer. J. Transl. Med. 2015, 13, 277. [Google Scholar] [CrossRef] [PubMed]
  150. Judge, S.J.; Yanagisawa, M.; Sturgill, I.R.; Bateni, S.B.; Gingrich, A.A.; Foltz, J.A.; Lee, D.A.; Modiano, J.F.; Monjazeb, A.M.; Culp, W.T.N.; et al. Blood and tissue biomarker analysis in dogs with osteosarcoma treated with palliative radiation and intra-tumoral autologous natural killer cell transfer. PLoS ONE 2020, 15, e0224775. [Google Scholar] [CrossRef] [PubMed]
  151. Denman, C.J.; Senyukov, V.V.; Somanchi, S.S.; Phatarpekar, P.V.; Kopp, L.M.; Johnson, J.L.; Singh, H.; Hurton, L.; Maiti, S.N.; Huls, M.H.; et al. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells. PLoS ONE 2012, 7, e30264. [Google Scholar] [CrossRef]
Figure 1. Schematic presentation of disease progression of HGSOC: HGSOCs likely consist of PDSLCs and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs contributing to disease progression and tumor relapse.
Figure 1. Schematic presentation of disease progression of HGSOC: HGSOCs likely consist of PDSLCs and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs contributing to disease progression and tumor relapse.
Vaccines 12 00677 g001
Figure 2. Schematic presentation of generation and characteristics of supercharging NK cells. NK cells (NK cells (0.5 × 106 cells/2 mL)) are purified from the peripheral blood of human donors and are treated with a combination of IL-2 (1000 U/mL) and anti-CD16mAb (3 µg/mL) overnight. Activated NK cells are then co-cultured with osteoclasts and probiotic bacteria (1:2:4: osteoclasts: NK: PB). Media were refreshed every three days for an average of 27–36 days. Supercharged NK cells exhibit significantly increased functional activity in comparison to primary NK cells.
Figure 2. Schematic presentation of generation and characteristics of supercharging NK cells. NK cells (NK cells (0.5 × 106 cells/2 mL)) are purified from the peripheral blood of human donors and are treated with a combination of IL-2 (1000 U/mL) and anti-CD16mAb (3 µg/mL) overnight. Activated NK cells are then co-cultured with osteoclasts and probiotic bacteria (1:2:4: osteoclasts: NK: PB). Media were refreshed every three days for an average of 27–36 days. Supercharged NK cells exhibit significantly increased functional activity in comparison to primary NK cells.
Vaccines 12 00677 g002
Figure 3. Hypothetical presentation of effective therapeutics for ovarian cancer. Chemotherapy alone could target well-differentiated tumors, but not PDSCSs, which could result in tumor recurrence. sNK cells induce killing against both PDSLCs and well-differentiated tumors and induce the differentiation of PDSLCs. A combination of sNK and chemotherapy could be the most effective strategy to eliminate the heterogeneous population of ovarian tumors: sNK therapies could help to kill both PDSLCs and well-differentiated tumors and induce the differentiation of PDSLCs, which could then allow chemotherapy to clean well-differentiated and NK-induced differentiated tumors.
Figure 3. Hypothetical presentation of effective therapeutics for ovarian cancer. Chemotherapy alone could target well-differentiated tumors, but not PDSCSs, which could result in tumor recurrence. sNK cells induce killing against both PDSLCs and well-differentiated tumors and induce the differentiation of PDSLCs. A combination of sNK and chemotherapy could be the most effective strategy to eliminate the heterogeneous population of ovarian tumors: sNK therapies could help to kill both PDSLCs and well-differentiated tumors and induce the differentiation of PDSLCs, which could then allow chemotherapy to clean well-differentiated and NK-induced differentiated tumors.
Vaccines 12 00677 g003
Table 1. Characteristic differences of primary vs. super-charged NK cells.
Table 1. Characteristic differences of primary vs. super-charged NK cells.
Primary IL-2-Activated NK CellsSupercharged NK CellsReferences
Lysis of PDSLCs+++++++ (****)[86,128,130]
Effector function associated gene-expression: IRF1, JUN, STAT1, H1F1A+++++ (***)[130]
ADCC against tumors+++++ (**)[132]
Cell survival6–9 days 27–36 days[130]
Cell survival associated gene-expression: STAT2, IRF9++++ (***)[130]
Cell expansion+/−++++ (****)[130]
Protein expression of cytotoxic granules+++ (**)[130]
Activating receptors surface expression++++++ (****)[130]
Inhibitory receptors surface expression levels +++ (***)+[130]
Stage of cell cycle% of cells: G1 > S > G2M% of cells: G2M > S > G1[130]
Majority of cells in sNK are highly proliferative
NK supernatant mediated tumor differentiation++++++ (****)[86,128,130]
Selection and expansion of CD8+ T cells (in vivo and in vitro)++++ (***)[129,135]
In vivo tumor growth and metastasis inhibition+++++ (****)[86,135]
PDSLCs: poorly-differentiated-cancer-stem-like-cells; ADCC: antibody-dependent cell cytotoxicity; Cytotoxic granules: granzyme B, cathepsin C, and perforin-1; + (1–2 fold), ++ (2–4 fold), +++ (3–4 fold), ++++ (4–6 fold), +++++ (>6 fold). **** (p value < 0.0001), *** (p value < 0.001), ** (p value 0.001–0.01).
Table 2. Clinical trial of NK cell-based ovarian cancer immunotherapy.
Table 2. Clinical trial of NK cell-based ovarian cancer immunotherapy.
Treatment ApproachClinical Trial StageClinicalTrials.gov Identifier
Allogeneic IV NK + IL-2 (IV)CompletedNCT01105650
Haploidentical NK + IL-2 + indoleamine-2,3-dioygenas (IDO) (IP)CompletedNCT02118285
Allogeneic NK + IL-2 (IP)CompletedNCT03213964
Cryosurgery + NKCompletedNCT02849353
TROP2-CAR IL-15 Transduced CB-NK (IP)In progressNCT05922930
Anti-mesothelin CAR-NKIn progressNCT03692637
Autologous activated NK (IV)In progressNCT03634501
Ex vivo generated UCB-derived allogeneic NK + IL-2 (IP)In progressNCT03539406
Cytokine-induced NK cells + radiofrequency ablationIn progressNCT02487693
UCB: umbilical cord blood-derived; IV: intravenous; IP: intraperitoneal; CB: cord blood.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kaur, K.; Sanghu, J.; Memarzadeh, S.; Jewett, A. Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas. Vaccines 2024, 12, 677. https://doi.org/10.3390/vaccines12060677

AMA Style

Kaur K, Sanghu J, Memarzadeh S, Jewett A. Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas. Vaccines. 2024; 12(6):677. https://doi.org/10.3390/vaccines12060677

Chicago/Turabian Style

Kaur, Kawaljit, Jashan Sanghu, Sanaz Memarzadeh, and Anahid Jewett. 2024. "Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas" Vaccines 12, no. 6: 677. https://doi.org/10.3390/vaccines12060677

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop