Anda di halaman 1dari 5

Leukemia & Lymphoma

ISSN: 1042-8194 (Print) 1029-2403 (Online) Journal homepage: https://www.tandfonline.com/loi/ilal20

Nivolumab induces dynamic alterations in CD8


T-cell function and TIM-3 expression when used
to treat relapsed acute myeloid leukemia after
allogeneic stem cell transplantation

Eric Wong, Joanne Davis, Rachel Koldej, Jeff Szer, Andrew Grigg & David
Ritchie

To cite this article: Eric Wong, Joanne Davis, Rachel Koldej, Jeff Szer, Andrew Grigg & David
Ritchie (2019): Nivolumab induces dynamic alterations in CD8 T-cell function and TIM-3 expression
when used to treat relapsed acute myeloid leukemia after allogeneic stem cell transplantation,
Leukemia & Lymphoma, DOI: 10.1080/10428194.2019.1648803

To link to this article: https://doi.org/10.1080/10428194.2019.1648803

Published online: 07 Aug 2019.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ilal20
LEUKEMIA & LYMPHOMA
https://doi.org/10.1080/10428194.2019.1648803

LETTER TO THE EDITOR

Nivolumab induces dynamic alterations in CD8 T-cell function and TIM-3


expression when used to treat relapsed acute myeloid leukemia after
allogeneic stem cell transplantation
Eric Wonga,b,c,d, Joanne Davisb, Rachel Koldejb, Jeff Szera,d, Andrew Griggc,d and David Ritchiea,b,d
a
Department of Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia;
b
Australian Cancer Research Foundation Translation Research Laboratory, Sydney, Australia; cDepartment of Clinical Oncology and
Olivia Newton-John Cancer Research Institute, Austin Hospital, Heidelberg, Australia; dDepartment of Medicine, Dentistry and Health
Sciences, University of Melbourne, Melbourne, Australia

ARTICLE HISTORY Received 30 April 2019; revised 3 July 2019; accepted 15 July 2019

Relapse of acute myeloid leukemia (AML) after allogeneic tachycardia syndrome (POTS) which was presumed to be
hematopoietic stem cell transplantation (alloSCT) is a immune-mediated. After eight doses of nivolumab,
major cause of mortality. Programmed death-1 (PD-1) repeat bone marrow biopsy demonstrated an increase in
and its cognate ligand PD-L1 may contribute to post- blast percentage to 17% and treatment was
alloSCT immune escape by repressing the CD8 T-cell discontinued.
mediated graft-versus-tumor (GVT) effect [1]. Blockade of Peripheral blood samples were obtained prior to nivo-
PD-1/PD-L1 interactions post-alloSCT is being investi- lumab and at regular intervals while on treatment. To
gated as a therapy for relapse, albeit with caution due to examine immune cell phenotypic and functional altera-
the risk of graft-versus-host disease (GVHD). Here, we tions following nivolumab treatment, mononuclear cells
describe the dynamics of T-cell phenotype and function were isolated by density gradient centrifugation and
in a patient who received nivolumab for AML relapsing stained with monoclonal antibodies (BD Biosciences,
after alloSCT. Franklin Lakes, NJ) including BD Horizon BUV395 conju-
A 43-year-old man with myelodysplastic syndrome gated anti-CD3, BV510 anti-CD4, BUV805 anti-CD8, BB515
(MDS) developed secondary AML with adverse prognosis anti-CD45RA, PE anti-CCR7, PE-Cy7 anti-HLA-DR, BUV496
features including a complex monosomal karyotype and anti-CD48, BV786 anti-PD1, BV421 anti-TIM3, Alexa Fluor
three TP53 coding region mutations. Induction chemo- 647 anti-LAG3, and fixable viability stain 780. Intracellular
therapy achieved complete morphologic remission from cytokine production was ascertained after four hours of
AML but persistent dysplasia consistent with the underly- in-vitro stimulation at 37  C with anti-CD2/3/28 microbe-
ing MDS. The patient proceeded to alloSCT from an ads (Miltenyi Biotec, Bergisch Gladbach, Germany) in the
HLA-matched unrelated donor with non-myeloablative presence of protein transport inhibitors (BD GolgiStop
conditioning due to severe hepatic steatosis. GVHD and GolgiPlug, BD Biosciences, Franklin Lakes, NJ) fol-
prophylaxis is comprised of in vivo T-cell depletion with lowed by staining with BV650 anti-IL2, APC anti-TNFa,
thymoglobulin in combination with ciclosporin and short and BV786 anti-IFNc. Samples were analyzed using an
course methotrexate. Bone marrow biopsy at day 30 LSRFortessa flow cytometer (BD Biosciences, Franklin
demonstrated complete morphologic remission. Relapse Lakes, NJ) and FlowJo software (Tree Star Inc., Ashland,
of AML occurred at 97 days post-alloSCT with 23% bone OR). Comparisons were made with healthy
marrow blasts. Immunosuppression was ceased without patient controls.
any ensuing GVHD. T-cell chimerism at relapse was 69% At post-transplant relapse prior to treatment with
donor origin. He was enrolled on a clinical trial of nivolu- nivolumab, CD8 T-cells is comprised of CD45RA-CCR7-T
mab for relapsed hematological malignancies after effector memory cells (Tem; 69.3%) and CD45RA þ CCR7-
alloSCT (ClinicalTrials.gov identifier: NCT03146468) and T effector cells (Teff; 24.9%) with a paucity of T naïve
received nivolumab 3 mg/kg every 2 weeks commencing cells (Tn; 0.8%) as expected at this early time point post-
120 days post-alloSCT. Disease re-assessment after four alloSCT. CD8 T-cells demonstrated increased expression
doses demonstrated reduction in blast count to 12%. of activation markers HLA-DR and CD38 compared with
There was no evidence of classical acute or chronic controls (HLA-DR 7.9% vs. 0.5%, p< .01; CD38 1.9% vs.
GVHD; however, he developed postural orthostatic 0.5%, p< .01). There was greater expression of PD-1, TIM-

CONTACT Eric Wong Eric.wong@mh.org.au Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Grattan Street,
Parkville, Victoria 3050, Australia
ß 2019 Informa UK Limited, trading as Taylor & Francis Group
2 E. WONG ET AL.

Figure 1. PD-1, TIM-3, and LAG-3 expression on CD8 T-cells in a healthy donor (A–C) and the patient case at the time of AML
relapse prior to treatment with nivolumab (D–F). There was a progressive increase in TIM-3 expression over time, with greatest
expression at the time of disease progression despite nivolumab (H–J). There was an initial increase in TNFa production with nivo-
lumab treatment however this was transient (K). At disease progression despite nivolumab treatment, there was upregulation of
TIM-3 expression and repression of TNFa production to baseline levels.
NIVOLUMAB AFTER ALLOGENEIC TRANSPLANT 3

3, and LAG-3 by CD8 T-cells at baseline compared with at the time of relapse post-alloSCT [1]. Furthermore, the
controls (PD-1 48.8% vs. 5.9%; TIM-3 19% vs. 5.5%; LAG-3 most impressive responses to PD-1 inhibitors either pre-
7.4% vs. 0.1%, all p< .01) (Figure 1(A–F)), and this was transplant or post-alloSCT are seen in patients with
seen in both CD8 Teff and Tem. Despite the increased Hodgkin lymphoma where upregulation of PD-L1 is an
expression of co-inhibitory receptors, CD8 T-cells intrinsic part of the tumor biology mediated by amplifica-
remained functionally competent and demonstrated tion of chromosome 9p24.1 [6].
increased IFNc (3.2% vs. 0.83%, p¼ .03) and IL-2 produc- Consistent with the early onset of clinical responses to
tion (5.6% vs. 0.8%, p¼ .004) and similar TNFa production nivolumab post-alloSCT, there was an early increase in
(2.6% vs. 1.4%, p¼ .3) compared with controls. CD8 T-cell TNFa production at seven days following nivo-
Following nivolumab treatment, there was a shift in lumab. Following initial response, leukemia progression
CD8 T-cell memory phenotype from Tem to Teff by day 7 was accompanied by an increase in TIM-3 expression and
together with an increase in TNFa production (5.7% vs. a decline in cytokine production, likely representing a
2.6%). There was no similar pattern in IFNc or IL2 produc- ‘true’ exhausted state. The upregulation of TIM-3 as an
tion over time. This shift in T-cell memory phenotype alternative inhibitory pathway may represent a mechan-
and increased cytokine production mirrored the onset of
ism of acquired resistance to PD-1 blockade, and has
the patient’s POTS symptoms, suggesting that this phe-
been reported in solid tumors [7]. In a murine model of
nomenon may have been immunologically driven. The
lung carcinoma, TIM-3 expression was upregulated with
increase in T-cell function was associated with a reduc-
increasing duration of PD-1 blockade. Therapeutic block-
tion in marrow blasts. At subsequent leukemia progres-
ade of TIM-3 at the time of acquired PD-1 resistance
sion despite continued nivolumab therapy, CD8 T-cells
achieved tumor responses indicating that loss of T-cell
demonstrated increased expression of TIM-3 compared
efficacy may be regained by sequential PD-1 followed by
with baseline (27% vs. 20%) (Figure 1(G–J)) which was
TIM3 inhibition [7]. Interestingly, there was no observed
accompanied by a decline in TNFa production to baseline
levels (Figure 1(K)), and a fall in IL-2 production at pro- change in IFNc or IL-2 production with nivolumab treat-
gression to below baseline (3.5% vs. 5.6%), suggesting ment, suggesting that checkpoint blockade post-alloSCT
that upregulation of TIM3 and repression of T-cell func- may not universally enhance T-cell functionality in a man-
tion may contribute to acquired resistance to PD-1 block- ner analogous to that observed in healthy donor T-cells
ade after alloSCT. in vitro [8]. Further analysis in a larger number of patients
This case report highlights several interesting findings. will be required to examine this hypothesis.
We demonstrate that nivolumab therapy post-alloSCT is In conclusion, we demonstrate that nivolumab
able to achieve leukemia response, albeit transient, in a induced a potent, if transient, GVT response in highly
patient with aggressive AML for which a non-augmented aggressive AML. Although PD-1 was upregulated prior to
GVT was insufficient. The use of checkpoint inhibitors to nivolumab therapy, CD8 T-cells remained functionally
treat relapsed AML after alloSCT has been described in intact and this was augmented by nivolumab therapy.
case series. In a phase I trial of CTLA4 inhibition post- Upregulation of TIM-3 was accompanied by reduced T-
alloSCT, there were five complete responses out of 18 cell function at leukemia progression, and may represent
patients with AML or MDS relapsing after alloSCT [2]. a mechanism of acquired resistance to PD-1 blockade
Albring et al. described three patients with relapsed AML post-alloSCT.
after alloSCT who received treatment with low dose nivo-
lumab, of which two patients achieved partial or com-
plete responses [3]. Disclosure statement
The proportion of PD-1 expressing CD8þ T-cells was The clinical trial was designed solely by the investigators
increased at the time of AML relapse compared with without input from Bristol-Myers Squibb, who also did not
healthy controls. Despite this phenotype, CD8 T-cells have any input into the analysis of results or preparation of
remained functionally competent with preserved cytokine the final manuscript. Bristol-Myers Squibb did not provide
production. Schnorfeil et al. reported preserved T-cell any other financial support to any of the authors.
function despite upregulation of PD-1 at the time of AML
relapse, suggesting that CD8 T-cell upregulation of PD-1
at relapse is not sufficient to indicate T-cell exhaustion Funding
[4]. PD-1 is upregulated by CD8 T-cells in response to The work described in this manuscript pertains to a clinical
activation and may not indicate exhaustion per se [5]. trial which was funded by Bristol-Myers Squibb.
Despite this, the biological rationale to pursue checkpoint
blockade in AML and other hematological malignancies
relapsing post-alloSCT remains, and response to this ther-
apy may be more closely linked to tumor expression of References
PD-L1 rather than a PD-1 positive T-cell phenotype. PD- [1] Norde WJ, Maas F, Hobo W, et al. PD-1/PD-L1 interactions
L1 has been described to be upregulated by AML blasts contribute to functional T-cell impairment in patients who
4 E. WONG ET AL.

relapse with cancer after allogeneic stem cell transplantation. [5] Legat A, Speiser DE, Pircher H, et al. Inhibitory receptor
Cancer Res. 2011;71:5111–5122. expression depends more dominantly on differentiation and
[2] Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for activation than “Exhaustion” of human CD8 T cells. Front
patients with relapse after allogeneic transplantation. N Engl Immunol. 2013;4:455.
J Med. 2016;375:143–153. [6] Haverkos BM, Abbott D, Hamadani M, et al. PD-1 blockade
[3] Albring JC, Inselmann S, Sauer T, et al. PD-1 checkpoint for relapsed lymphoma post-allogeneic hematopoietic cell
transplant: high response rate but frequent GVHD. Blood.
blockade in patients with relapsed AML after allogeneic stem
2017;130:221–228.
cell transplantation. Bone Marrow Transplant. 2017;52:
[7] Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to
317–320. therapeutic PD-1 blockade is associated with upregulation of
[4] Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. T cells are
alternative immune checkpoints. Nat Commun. 2016;7:10501.
functionally not impaired in AML: increased PD-1 expression [8] Wang C, Thudium KB, Han M, et al. In vitro characterization
is only seen at time of relapse and correlates with a shift of the anti-PD-1 antibody nivolumab, BMS-936558, and in
towards the memory T cell compartment. J Hematol Oncol. vivo toxicology in non-human primates. Cancer Immunol
2015;8:93. Res. 2014;2:846–856.

Anda mungkin juga menyukai