Elsevier

Cytotherapy

Volume 13, Issue 1, January 2011, Pages 98-107
Cytotherapy

A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer

https://doi.org/10.3109/14653249.2010.515582Get rights and content

Abstract

Background

Natural killer (NK) cells derived from patients with cancer exhibit diminished cytotoxicity compared with NK cells from healthy individuals. We evaluated the tumor response and in vivo expansion of allogeneic NK cells in recurrent ovarian and breast cancer

Methods

Patients underwent a lymphodepleting preparative regimen: fludarabine 25 mg/m2 × 5 doses, cyclophosphamide 60 mg/kg × 2 doses, and, in seven patients, 200 cGy total body irradiation (TBI) to increase host immune suppression. An NK cell product, from a haplo-identical related donor, was incubated overnight in 1000 U/mL interleukin (IL)-2 prior to infusion. Subcutaneous IL-2 (10 MU) was given three times/week × 6 doses after NK cell infusion to promote expansion, defined as detection of ≥100 donor-derived NK cells/μL blood 14 days after infusion, based on molecular chimerism and flow cytometry

Results

Twenty (14 ovarian, 6 breast) patients were enrolled. The median age was 52 (range 30–65) years. Mean NK cell dose was 2.16 × 107cells/kg. Donor DNA was detected 7 days after NK cell infusion in 9/13 (69%) patients without TBI and 6/7 (85%) with TBI. T-regulatory cells (Treg) were elevated at day +14 compared with pre-chemotherapy (P = 0.03). Serum IL-15 levels increased after the preparative regimen (P = < 0.001). Patients receiving TBI had delayed hematologic recovery (P = 0.014). One patient who was not evaluable had successful in vivo NK cell expansion

Conclusions

Adoptive transfer of haplo-identical NK cells after lymphodepleting chemotherapy is associated with transient donor chimerism and may be limited by reconstituting recipient Treg cells. Strategies to augment in vivo NK cell persistence and expansion are needed.

Introduction

Human natural killer (NK) cells are a subset of peripheral blood lymphocytes defined by the expression of CD56 or CD16 and the absence of T-cell receptor (CD3) (1., 2., 3., 4., 5., 6., 7., 8., 9., 10.). NK cells play a role in tumor surveillance and can recognize unhealthy cells with decreased expression of class I major histocompatibility complex (MHC) molecules, referred to as ‘loss of self’ (11,12). In a number of tumor types generally considered unresponsive to chemotherapy, a series of well-publicized trials at the National Cancer Institute (NCI, Bethesda, MD, USA) has documented anti-tumor effects in patients using adoptive cellular immunotherapy (ACT). (13,14). This approach involves harvesting mononuclear cells from patients via lymphapheresis, incubation of the cells ex vivo using high concentrations of the lymphokine interleukin (IL)-2, and administration of the expanded and IL-2-activated cells (lymphokine-activated killer, or LAK, cells) to the patient along with IL-2 administration. Early clinical trials showed modest clinical success using autologous LAK with high-dose IL-2 in lymphoma, melanoma and renal cancers, with the majority of cytotoxicity attributed to NK cells (15). The rationale for this study was the potent function of IL-2-activated allogeneic NK cells, compared with autologous NK cells, against ovarian and breast cancer (16., 17., 18., 19., 20.). We now understand that the failure of autologous NK therapy is partially because of the down-regulation of NK cell killing that occurs with recognition of self-class I MHC on tumor cells, making allogeneic cell transfer more attractive (11,12,21).

Murine models show that depletion of immune cells before ACT enhances the anti-tumor efficacy of transferred donor cells, with a direct correlation between the extent of lymphodepletion and in vivo anti-tumor effect of the transferred cells (22). Lymphodepletion has been shown to augment innate immunity by increasing exposure to homeostatic cytokines (IL-7 and IL-15), eliminating competing elements of the immune system (‘cytokine sinks’) and limiting the number of regulatory T lymphocytes (Treg) and myeloid-derived suppressor cells (23,24). Lymphodepletion followed by ACT has produced approximately 20% complete and partial responses in initial trials at the NCI, with responses occurring primarily in melanoma, renal cell cancers and non-Hodgkin lymphoma (15). A clinical trial assessing the safety and efficacy of related donor, HLA-haplo-identical, allogeneic NK-enriched peripheral blood cell infusion in patients with poor prognosis acute myeloid leukemia (AML) has been completed at the University of Minnesota (25). We learned that infusion of related donor haplo-identical allogeneic NK cell infusions is safe and that successful in vivo donor NK cell expansion, which correlates with efficacy in AML, requires a high-dose cyclophosphamide and fludarabine lymphodepleting preparative regimen (Hi-Cy/Flu). More recently, accumulating data in animal models suggest that further lymphodepletion may improve ACT persistence and efficacy (23,26). Dudley et al. (22) evaluated the efficacy and safety of adding total body irradiation (TBI) to a non-myeloablative chemotherapy preparative regimen in patients with metastatic melanoma. They reported an objective response rate of 50–70%. Additionally, they found increases in homeostatic cytokines IL-7 and IL-15, hypothesized to lead to the persistence, proliferation and activation of the adoptively transferred cells.

Based on the above findings, we evaluated the in vivo expansion and clinical efficacy of an adoptively transferred haplo-identical donor NK cell product in a solid tumor setting following a preparative regimen with and without total body irradiation (TBI).

Section snippets

Patient eligibility

Patients over the age of 18 years with refractory metastatic breast or ovarian cancer with adequate performance status, organ function [total bilirubin, Aspartate transaminase(AST)/Alanine transaminase(ALT) ≤ 5 times upper limits of normal, and creatinine < 2.0 mg/dL, or calculated creatinine clearance ≥50 mL/min for patients with creatinine levels above normal] and hematologic reserve (platelet count greater than 80,000/μL, hemoglobin level greater than 9 gm/dL, and an absolute neutrophil count

Characteristics of the NK cell products

The apheresis products contained a median of 9.5% CD56+ CD3 NK cells (range 4.0–29.6%) prior to manipulation. Following enrichment by CD3 depletion, the final products contained a median of 33% NK cells (range 23.1–55.5%), resulting in a median infused NK cell dose of 2.15 × 107 NK cells/kg (range 8.33 × 106−3.94 × 107). The products were significantly T-cell depleted after processing, with a median of 0.11% CD3+ cells (range 0.03–1.9%), giving a median infused T-cell dose of 5.59 × 104 CD3+

Discussion

We tested the use of adoptively transferred haplo-identical NK cells to treat patients with solid tumors. Following preparation with a Hi-Cy/Flu regimen, donor DNA was detectable 7 days after the NK cell infusion in 79% (15/19) of patients. Although most patients had high percentages of NK cells at day +7, the absolute lymphocyte count was essentially immeasurable at that time-point. After completion of the IL-2 course 14 days after NK cell infusions, the small numbers of donor NK cells found

Acknowledgments

We would like to thank Dixie Lewis and Megan Whitmore for their excellence in study coordination and patient care during this trial, and Giordi Orreggio for his skills in data management. Additionally, we thank Jessica Kuehn-Hajder, MD, for her radiologic expertise in this trial.

Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References (55)

  • CooleyS. et al.

    Successful haploidentical hematopoietic cell engraftment using a non-myeloablative preparative regimen including natural killer (NK) cells

    Blood

    (2008)
  • RosenbergS.A. et al.

    Adoptive cell therapy for the treatment of patients with metastatic melanoma

    Curr Opin Immunol

    (2009)
  • AraiS. et al.

    Infusion of the allogeneic cell line NK-92 in patients with advanced renal cell cancer or melanoma: a phase I trial

    Cytotherapy

    (2008)
  • ZhouQ. et al.

    Depletion of endogenous tumor-associated regulatory T cells improves the efficacy of adoptive cytotoxic T-cell immunotherapy in murine acute myeloid leukemia

    Blood

    (2009)
  • LutsiakM.E. et al.

    Inhibition of CD4(+)25+ T regulatory cell function implicated in enhanced immune response by low-dose cyclophosphamide

    Blood

    (2005)
  • BeyerM. et al.

    Reduced frequencies and suppressive function of CD4+CD25hi regulatory T cells in patients with chronic lymphocytic leukemia after therapy with fludarabine

    Blood

    (2005)
  • ZornE. et al.

    Combined CD4+ donor lymphocyte infusion and low-dose recombinant IL-2 expand FOXP3+ regulatory T cells following allogeneic hematopoietic stem cell transplantation

    Biol Blood Marrow Transplant

    (2009)
  • YokoyamaH. et al.

    Adoptively infused NK cells maintain their antitumor effects in vivo in the presence of cyclosporin A (CSA)

    Blood

    (2008)
  • YahataT. et al.

    Tumor lysis syndrome associated with weekly paclitaxel treatment in a case with ovarian cancer

    Gynecol Oncol

    (2006)
  • PiersonB. et al.

    Population dynamics of human activated killer cells in culture

    Biotech Bioeng

    (1994)
  • MillerJ.S. et al.

    Large scale ex vivo expansion and activation of human natural killer cells for autologous therapy

    Bone Marrow Transplant

    (1994)
  • PiersonB.A. et al.

    Natural killer cell proliferation is dependent on human serum and markedly increased utilizing an enriched supplemented basal medium

    J Hematother

    (1995)
  • PiersonB.A. et al.

    Production of human natural killer cells for adoptive immunotherapy using a computer-controlled stirred-tank bioreactor

    J Hematother

    (1996)
  • KarreK.

    Express yourself or die: peptides, MHC molecules, and NK cells

    Science

    (1995)
  • RosenbergS.A.

    Karnofsky Memorial lecture. The immunotherapy and gene therapy of cancer

    J Clin Oncol

    (1992)
  • CanevariS. et al.

    Regression of advanced ovarian carcinoma by intraperitoneal treatment with autologous T lymphocytes retargeted by a bispecific monoclonal antibody

    J Natl Cancer Inst

    (1995)
  • RosenbergS.A. et al.

    A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone

    N Engl J Med

    (1987)
  • Cited by (356)

    • IPSC-derived CAR-NK cells for cancer immunotherapy

      2023, Biomedicine and Pharmacotherapy
    View all citing articles on Scopus
    *

    Equal contributing fi rst authors.

    View full text