Increased mTORC2 pathway activation in lymph nodes of iMCD-TAFRO

Alexis D Phillips, Joseph J Kakkis, Patricia Y Tsao, Sheila K Pierson, David C Fajgenbaum, Alexis D Phillips, Joseph J Kakkis, Patricia Y Tsao, Sheila K Pierson, David C Fajgenbaum

Abstract

Idiopathic multicentric Castleman disease (iMCD) is a rare and life-threatening haematologic disorder involving polyclonal lymphoproliferation and organ dysfunction due to excessive cytokine production, including interleukin-6 (IL-6). Clinical trial and real-world data demonstrate that IL-6 inhibition is effective in 34-50% of patients. mTOR, which functions through mTORC1 and mTORC2, is a recently discovered therapeutic target. The mTOR inhibitor sirolimus, which preferentially inhibits mTORC1, has led to sustained remission in a small cohort of anti-IL-6-refractory iMCD patients with thrombocytopenia, anasarca, fever, renal dysfunction and organomegaly (iMCD-TAFRO). However, sirolimus has not shown uniform effect, potentially due to its limited mTORC2 inhibition. To investigate mTORC2 activation in iMCD, we quantified the mTORC2 effector protein pNDRG1 by immunohistochemistry of lymph node tissue from six iMCD-TAFRO and eight iMCD patients who do not meet TAFRO criteria (iMCD-not-otherwise-specified; iMCD-NOS). mTORC2 activation was increased in all regions of iMCD-TAFRO lymph nodes and the interfollicular space of iMCD-NOS compared with control tissue. Immunohistochemistry also revealed increased pNDRG1 expression in iMCD-TAFRO germinal centres compared with autoimmune lymphoproliferative syndrome (ALPS), an mTOR-driven, sirolimus-responsive lymphoproliferative disorder, and comparable staining between iMCD-NOS and ALPS. These results suggest increased mTORC2 activity in iMCD and that dual mTORC1/mTORC2 inhibitors may be a rational therapeutic approach.

Keywords: Castleman disease; TAFRO; autoimmune lymphoproliferative syndrome; iMCD; idiopathic multicentric Castleman disease; mTOR; mTORC2; pNDRG1.

Conflict of interest statement

D.C.F. has received research funding for the ACCELERATE registry (NCT02817997) from EUSA Pharma and consulting fees from EUSA Pharma, and Pfizer provides study drug with no associated research funding for the clinical trial of sirolimus (NCT03933904). D.C.F. has two provisional patents pending related to the diagnosis and treatment of iMCD. The remaining authors declare no competing interests.

© 2022 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Comparison of pNDRG1 staining across iMCD subtypes and autoimmune lymphoproliferative syndrome (ALPS). (A) Stained pNDRG1 for iMCD‐TAFRO patients (n = 6) and (B) iMCD‐not otherwise specified (iMCD‐NOS) patients (n = 8) compared with a control group of metastasis‐free lymph nodes (normal) (n = 7). Relative to normal lymph nodes, pNDRG1 positive area was significantly elevated in the interfollicular space (p = 0.005), germinal centre (p = 0.002) and mantle zone (p = 0.007) of iMCD‐TAFRO lymph nodes. pNDRG1 positive area was significantly elevated in the interfollicular space (p = 0.005) of iMCD‐NOS lymph nodes relative to normal lymph nodes, but there was no difference in staining in the germinal centres (p = 0.59) and mantle zones (p = 0.30). (C) Stained pNDRG1 area for iMCD‐TAFRO patients (n = 6) and iMCD‐NOS patients (n = 8) compared with ALPS lymph nodes (n = 8). There was a significant increase in positive pNDRG1 staining in iMCD‐TAFRO germinal centres relative to ALPS (p = 0.02), and no difference in the interfollicular space (p = 0.18) and mantle zone (p = 0.11). There was no difference in positive pNDRG1 staining in iMCD‐NOS relative to ALPS in the interfollicular space (p = 1.0), germinal centres (p = 1.0) and mantle zones (p = 1.0). *p <.05, **p <.01. D‐G, Representative images of pNDRG1 (brown) staining for a (D) normal lymph node (E) iMCD‐TAFRO (F) iMCD‐NOS and (G) ALPS lymph node. Haematoxylin counterstain provides a blue nuclear stain to assess cell and tissue morphology. Bar = 400 μm. (H) The strongest pNDRG1‐positive cells appear to have spindle‐shaped morphology resembling stromal cells. Bar = 100 μm.

References

    1. Iwaki N, Fajgenbaum DC, Nabel CS, et al. Clinicopathological analysis of TAFRO syndrome demonstrates a distinct subtype of HHV‐8‐negative multicentric Castleman disease. Am J Hematol. 2015;91(2):220‐226.
    1. Dispenzieri A, Fajgenbaum DC. Overview of Castleman disease. Blood. 2020;135(16):1353‐1364.
    1. van Rhee F, Voorhees P, Dispenzieri A, et al. International, evidence‐based consensus treatment guidelines for idiopathic multicentric Castleman disease. Blood. 2018;132(20):2115‐2124.
    1. Van Rhee F, Wong RS, Munshi N, et al. Siltuximab for multicentric Castleman’s disease: a randomized, double‐blind, placebo‐controlled trial. Lancet Oncol. 2014;15(9):966‐974.
    1. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960‐976.
    1. Arenas DJ, Floess K, Kobrin D, et al. Increased mTOR activation in idiopathic multicentric Castleman disease. Blood. 2020;135(19):1673‐1684.
    1. Fajgenbaum DC, Langan RA, Japp AS. Identifying and targeting pathogenic PI3K/AKT/mTOR signaling in IL‐6 blockade‐refractory idiopathic multicentric Castleman disease. J Clin Invest. 2019;129(10):4451‐4463.
    1. Bride KL, Vincent T, Smith‐Whitley K, et al. Sirolimus is effective in relapse/refractory autoimmune cytopenias: results of a prospective multi‐institutional trial. Blood. 2016;127(1):17‐28.
    1. García‐Martínez JM. mTOR complex 2 (mTORC2) controls hydrophobic motif phosphorylation and activation of serum‐and glucocorticoid‐induced protein kinase 1 (SGK1). Biochem J. 2008;146(3):375‐385.
    1. Tanaka K, Babic I, Nathanson D, et al. Oncogenic EGFR signaling activates an mTORC2‐NF‐kB pathway that promotes chemotherapy resistance. Cancer Discov. 2011;1(6):524‐538.
    1. Huang J, Wu S, Wu CL, Manning BD. Signaling events downstream of mammalian target of rapamycin complex 2 are attenuated in cells and tumors deficient for the tuberous sclerosis complex tumor suppressors. Cancer Res. 2009;69:6107‐6114.
    1. Fu W, Hall MN. Regulation of mTORC2 signaling. Genes (Basel). 2020;11(9):1045.
    1. Weiler M, Blaes J, Pusch S, et al. mTOR target NDRG1 confers MGMT‐dependent resistance to alkylating chemotherapy. Proc Natl Acad Sci USA. 2014;111(1):409‐414.
    1. Oh WJ, Jacinto E. mTOR complex 2 signaling and functions. Cell Cycle. 2011;10(14):2305‐2316.
    1. Kazyken D, Magnuson B, Bodur C, et al. AMPK directly activates mTORC2 to promote cell survival during acute energetic stress. Science Signaling. 2019;12(585)eaav3249.
    1. Voss MH, Gordon MS, Mita M. Phase 1 study of mTORC1/2 inhibitor sapanisertib (TAK‐228) in advanced solid tumours, with an expansion phase in renal, endometrial or bladder cancer. Br J Cancer. 2020;123:1590‐1598.
    1. Pierson SK, Khor JS, Ziglar J, et al. ACCELERATE: a patient‐powered natural history study design enabling clinical and therapeutic discoveries in a rare disorder. Cell Rep Med. 2020;1(9):100158.
    1. Nishimura Y, Fajgenbaum DC, Pierson SK, et al. Validated international definition of the thrombocytopenia, anasarca, fever, reticulin fibrosis, renal insufficiency, and organomegaly clinical subtype (TAFRO) of idiopathic multicentric Castleman disease. Am J Hematol. 2021;96(10):1241‐1252.
    1. Iwaki N, Fajgenbaum DC, Nabel CS, et al. Clinicopathologic analysis of TAFRO syndrome demonstrates a distinct subtype of HHV‐8‐negative multicentric Castleman disease. Am J Hematol. 2016;91(2):220‐226.
    1. Subbiah V, Brown RE, McGuire MF, et al. A novel immunomodulatory molecularly targeted strategy for refractory Hodgkin's lymphoma. Oncotarget. 2014;5(1):95‐102.
    1. Pai RL, Japp AS, Gonzalez M, et al. Type I IFN response associated with mTOR activation in the TAFRO subtype of idiopathic multicentric Castleman disease. JCI Insight. 2020;5(9):e135031.
    1. Pierson SK, Shenoy S, Oromendia AB, et al. Discovery and validation of a novel subgroup and therapeutic target in idiopathic multicentric Castleman disease. Blood Adv. 2021;5(17):3445‐3456.
    1. Fajgenbaum DC, Uldrick TS, Bagg A, et al. International, evidence‐based consensus diagnostic criteria for HHV‐8‐negative/idiopathic multicentric Castleman disease. Blood. 2017;129(12):1646‐1657.
    1. Pierson S, Stonestrom AJ, Shilling D, et al. Plasma proteomics identifies a ‘chemokine storm’ in idiopathic multicentric Castleman disease. Am J Hematol. 2018;93(7):902‐912.

Source: PubMed

3
Subskrybuj