Autologous stem-cell transplantation (ASCT) for multiple myeloma continues to confer benefit in the “novel agent” era [1]. However, most patients relapse, risk stratification is inadequate and optimal post-ASCT maintenance therapy strategies remain unclear. Relapses are heralded by the emergence of IL-10-secreting regulatory myeloid cells (immunosuppressive macrophages and dendritic cells) and dysfunctional (exhausted/senescent) T cells expressing programmed death −1 (PD-1) and T-cell immunoglobulin and ITIM domain (TIGIT) receptors [2–5]. Moreover, IL-10-induced CD8+ regulatory T cells impede antigen-specific effector responses [6,7]. Infiltrating myeloid cells secrete versican (VCAN), a large matrix proteoglycan with immunosuppressive/ tolerogenic activities [8–10]. ADAMTS protease-mediated VCAN cleavage generates versikine, a bioactive proteolytic fragment (“matrikine”), that regulates BATF3-dendritic cell (BATF3-DC) differentiation [11]. BATF3-DC are a crucial DC subset that controls CD8+ abundance in the tumor microenvironment through enhanced CD8+ effector cross-priming (BATF3-DC are the most efficient DC subset in tumor antigen cross-presentation) as well as through chemokine-mediated CD8+ infiltration into tumor sites [12].
Immune reconstitution post-ASCT provides a favorable milieu for the institution of immunotherapies that aim to enhance effector function and control or eradicate minimal residual myeloma [4]. In this work, we sought to answer two questions: Firstly, does VCAN proteolysis correlate with CD8+ infiltration in the post-ASCT marrow microenvironment? Secondly, does VCAN proteolysis status carry prognostic significance in the post-ASCT setting?
Bone marrow core biopsies were collected with informed consent under a UW-Madison Institutional Review Board-approved protocol (HO07403). VCAN proteolysis status was determined through immunohistochemical staining for the neo-epitope DPEAAE that is exposed through ADAMTS-mediated proteolytic VCAN cleavage at the Glu441- Ala442 bond of the VCAN-V1 isoform [13]. DPEAAE constitutes the C-terminal end of the bioactive VCAN fragment, versikine [13]. Antigen retrieval was carried out in EDTA buffer (CD8 detection) or citrate (DPEAAE). Primary antibodies included anti-DPEAAE (PA1–1748A; Thermo Fisher, Waltham, MA) and anti-CD8 (c4–0085-80; eBioscience, San Diego, CA). Core biopsy anti-DPEAAE staining intensity was scored for each sample by at least three observers, including a pathologist (AMC), blinded to clinical parameters. Stained slides were examined using Olympus BX43 microscope with an attached Olympus DP73 digital camera (Olympus, Waltham, MA). Immunostaining for anti-DPEAAE was assessed by scoring staining intensity (1 for low/weak, 2 moderate and 3 for strong/intense staining). For CD8+ detection, the number of CD8+ T cells per hpf was calculated using a single area at 400X magnification (10X ocular with a 40X objective). Baseline and post-ASCT characteristics between the low and moderate/high VCAN proteolysis groups were compared using chi-square test, Fisher’s exact test or t-test as appropriate. Correlation of VCAN proteolysis status with CD8+ T cell count per hpf was performed using ANOVA and logistic regression. OS and PFS between the two groups were estimated using Kaplan-Meier method. Cox regression analyses were used to correlate factors with OS and PFS using VCAN proteolysis status as the main effect and hazard ratios (HR) and 95% confidence intervals (CI) were obtained. Logistic regression analyses were used to estimate predictors of 2-year OS and PFS and odds ratios (OR) with 95% CI were ascertained. Data were analyzed using SPSS version 21 (SPSS Inc, Chicago, IL), and p<0.05 was considered statistically significant.
Bone marrow biopsies from 35 myeloma patients collected at Day 90–100 post-ASCT were analyzed. Table 1 provides patient baseline characteristics. The median age was 61 years (range 24–74) and 63% were males. Twenty four percent had high-risk cytogenetics, and 18% had very good partial response or better (≥VGPR) at the immediate pre-transplant evaluation. All patients received high dose melphalan at 200 mg/m2 for conditioning. Disease evaluation was done at day 90–100 which showed that 73% of patients were in ≥VGPR. Most patients received maintenance therapy (63%) with no significant differences between VCAN-proteolysis classification groups (p= 0.455, Table 1). Anti-DPEAAE staining was robust despite prior decalcification of samples, a condition known to adversely impact detection of other immune biomarkers, such as PD-L1, by immunohistochemistry. VCAN proteolysis-low intensity (1+) was present in 18 (51%) patients, while VCAN proteolysis-high intensity (2+ and 3+) was present in 17 (49%) patients (2+ intensity= 11 patients and 3+ intensity= 6 patients). Representative staining patterns are given in Fig. 1A. Core biopsies were stained for the effector T cell marker CD8 and CD8+ counts per high-power field (hpf) were correlated with the VCAN proteolysis status (Fig. 1A). There was a statistically significant correlation between VCAN-proteolysis intensity and CD8+ T cell infiltration (VCAN proteolysis intensity 1+: CD8+ count per hpf mean, SD= 17.7 (13.1); VCAN proteolysis intensity 2+: CD8+ count per hpf mean, SD= 27 (14.8); VCAN proteolysis intensity 3+: CD8+ count per hpf mean, SD= 67.3 (43.1); p<0.001) (Fig. 1B). On univariate logistic regression, intense VCAN proteolysis (2–3+) was associated with higher CD8+ T cell count (OR, 1.06, 95% CI 1.01–1.12; p=0.027).
Table 1:
Baseline and post-transplant characteristics of patients.
Characteristics | Total (n=35) | VCAN proteolysis | p value | |
---|---|---|---|---|
Low (1+) (n=18) | Moderate/High (2–3+) (n=17) | |||
Age in years, median (range) | 61 (24–74) | 60 (24–74) | 61 (46–73) | 0.369 |
Male gender, n (%) | 22 (63) | 13 (72) | 9 (53) | 0.305 |
Caucasian race, n (%) | 33 (94) | 17 (94) | 16 (94) | 0.367 |
Isotype, n (%) | ||||
IgG lambda | 4 (11) | 3 (17) | 1 (6) | 0.285 |
IgA lambda | 2 (6) | 1 (6) | 1 (6) | |
IgG kappa | 16 (46) | 9 (50) | 7 (6) | |
IgA kappa | 4 (11) | 0 | 4 (23.5) | |
Lambda | 3 (9) | 1 (6) | 2 (12) | |
Kappa | 6 (17) | 4 (22) | 2 (12) | |
ISS stage, n (%) | ||||
I | 9 (26) | 6 (33) | 3 (18) | 0.177 |
II | 11 (31) | 7 (39) | 4 (23.5) | |
III | 15 (43) | 5 (28) | 10 (59) | |
Cytogenetics, n (%) | ||||
Standard risk | 26 (76.5) | 15 (88) | 11 (65) | 0.225 |
High risk | 8 (23.5) | 2 (12) | 6 (35) | |
Induction regimen, n (%) | ||||
VRD | 12 (34) | 7 (39) | 5 (29) | 0.311 |
CyBorD | 4 (11) | 2 (11) | 2 (11) | |
Rd | 8 (23) | 4 (22) | 4 (23.5) | |
Vd | 5 (14) | 4 (22) | 1 (6) | |
Others | 6 (17) | 1 (6) | 5 (29) | |
Pre-transplant response, n (%) | ||||
sCR/CR | 3 (9) | 1 (6) | 2 (12) | 0.907 |
nCR/VGPR | 3 (9) | 2 (11) | 1 (6) | |
PR | 22 (63) | 12 (67) | 10 (59) | |
SD | 2 (6) | 1 (6) | 1 (6) | |
PD | 5 (14) | 2 (11) | 3 (18) | |
Post-transplant response, n (%) | ||||
sCR/CR | 16 (46) | 11 (61) | 5 (29) | 0.071 |
nCR/VGPR | 10 (27) | 6 (33) | 4 (23.5) | |
PR | 3 (9) | 0 | 3 (18) | |
SD | 2 (6) | 0 | 2 (12) | |
PD | 4 (11) | 1 (6) | 3 (18) | |
Maintenance therapy, n (%) | ||||
Lenalidomide | 11 (31) | 7 (39) | 4 (23.5) | 0.455 |
Others | 11 (31) | 6 (33) | 5 (29) | |
None | 13 (37) | 5 (28) | 8 (47) | |
Follow-up in months, median (range) | 32 (7–116) | 36 (8–116) | 13 (7–80) | 0.088 |
Abbreviations: VRD (Bortezomib, Lenalidomide, Dexamethasone), CyBorD (Cyclophosphamide, Bortezomib, Dexamethasone), Rd (Lenalidomide, Dexamethasone), Vd (Bortezomib, Dexamethasone), sCR (stringent complete remission), CR (complete remission), nCR (near complete remission), VGPR (very good partial response), PR (partial remission), SD (stable disease), PD (progressive disease).
Figure 1.
(A), Representative photomicrographs of post-ASCT bone marrow biopsies stained with an antibody detecting VCAN proteolysis (anti-DPEAAE neoepitope) (top) and an anti-CD8 antibody (bottom). hpf= high-power field. (B) Box-and-whisker plots demonstrating the distribution of CD8+ count per hpf according to VCAN proteolysis intensity. (C) Kaplan-Meier curves demonstrating overall survival (OS, p= 0.003) (left) and progression-free survival (PFS, p=0.054) (right) according to VCAN proteolysis intensity: low (1+) and moderate/high (2+ and 3+ intensity combined).
Outcomes were compared between patients with low VCAN proteolysis intensity (1+) and a combined group comprising of patients with moderate and high VCAN proteolysis (2+ and 3+ combined). Overall response rate (ORR) was 83% and 46% patients achieved complete remission. ORR and the depth of response, while superior in patients with low VCAN proteolysis intensity, were not statistically significant (p=0.071, Table 1). Low VCAN proteolysis compared to moderate/high VCAN proteolysis was associated with better OS (median not reached vs. 13 months, 95% CI 10–16, p=0.003) and a trend for better PFS (median 56 months; 95% CI 26–87 vs. 10 months, 95% CI 1–18; p=0.054) (Fig. 1C). In Cox regression analyses adjusted for age and gender, moderate/high VCAN proteolysis compared to low VCAN proteolysis predicted poor OS (HR 4.87, 95% CI 1.33–17.82; p=0.017) and a similar trend was seen for PFS although it was not statistically significant (HR 2.29, 95% CI 0.92–5.68; p=0.074). Higher CD8+ count as a continuous variable was associated with poor OS (HR 1.03, 95% CI 1.01–1.05; P=0.002). In multivariate Cox regression model including VCAN proteolysis status, CD8+ count and cytogenetic risk group, moderate/high VCAN proteolysis compared to low VCAN proteolysis remained independent predictor of poor OS (HR 5.07, 95% CI 1.04–24.76; p=0.045). Patients with low VCAN proteolysis compared to moderate/high VCAN proteolysis had better 2-year PFS (72% vs. 29%, p=0.018) and 2-year OS (83% vs. 35%, p=0.006). In logistic regression analyses adjusted for age and gender, low VCAN proteolysis compared to moderate/high VCAN proteolysis predicted higher 2-year PFS (OR 6.80, 95% CI 1.41–32.76; p=0.017) and 2-year OS (OR 7.64, 95% CI 1.50–39.07; p=0.015). In age- and gender-adjusted multivariate logistic regression model including VCAN proteolysis status, CD8+ count and cytogenetic risk group, low VCAN proteolysis compared to moderate/high VCAN proteolysis independently predicted better 2-year OS (OR 7.77, 95% CI 1.06–56.71; p=0.043) and 2-year PFS (OR 8.48, 95% CI 1.09–66.09; p=0.041).
We present the first set of data ascribing prognostic significance to the VCAN proteolysis immunoregulatory pathway in myeloma. We observed the somewhat paradoxical association between intense VCAN proteolysis and high CD8+ T cell infiltration with dismal post-ASCT survival. These observations support a model in which immunosuppressive myeloid cells (macrophages and regulatory DC) secrete tolerogenic VCAN. Subsequent proteolysis of accumulated VCAN releases versikine. Versikine in turn promotes CD8+ infiltration (likely through BATF3-DC chemokine networks [12]). Intramedullary CD8+ effectors become dysfunctional and/or frankly immunoregulatory at the tumor site [2–7], thus perpetuating immunosuppressive loops and sustaining conditions favorable for relapse. Multiparametric analyses suggested negative effects of VCAN proteolysis independent of CD8+ T cell status, alluding to yet unexplored interactions between intrinsically high-risk disease and tumor matrix remodeling.
The detection of VCAN accumulation and turnover thus indicates profound immunosuppression in the post-ASCT marrow microenvironment and pinpoints those patients at risk for relapse and early death. The dismal outcomes associated with VCAN biomarker positivity may be rationally overcome through cutting-edge immunotherapies such as anti-TIGIT checkpoint inhibition [2], dendritic cell vaccines [14] or myeloid reprogramming therapies [3,15]. At the time of writing, VCAN proteolysis detection has been incorporated as an ancillary study in a Phase II autologous DC/myeloma cell fusion vaccination protocol (NCT02728102).
ACKNOWLEDGEMENTS
This work was supported by the Leukemia and Lymphoma Society (6551–18), the American Cancer Society (127508-RSG-15–045-01-LIB), the UWCCC Trillium Fund for Myeloma Research and the NIH (P30CA014520).
Footnotes
CONFLICTS
None of the authors has any relevant conflicts to declare.
REFERENCES
- 1.Dhakal B, Szabo A, Chhabra S, et al. . Autologous Transplantation for Newly Diagnosed Multiple Myeloma in the Era of Novel Agent Induction: A Systematic Review and Meta-analysis. JAMA Oncol 2018;4:343–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Guillerey C, Harjunpaa H, Carrie N, et al. . TIGIT immune checkpoint blockade restores CD8(+) T-cell immunity against multiple myeloma. Blood 2018;132:1689–1694. [DOI] [PubMed] [Google Scholar]
- 3.Minnie SA, Kuns RD, Gartlan KH, et al. . Myeloma escape after stem cell transplantation is a consequence of T-cell exhaustion and is prevented by TIGIT blockade. Blood 2018;132:1675–1688. [DOI] [PubMed] [Google Scholar]
- 4.Chung DJ, Pronschinske KB, Shyer JA, et al. . T-cell Exhaustion in Multiple Myeloma Relapse after Autotransplant: Optimal Timing of Immunotherapy. Cancer Immunol Res 2016;4:61–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Suen H, Brown R, Yang S, et al. . Multiple myeloma causes clonal T-cell immunosenescence: identification of potential novel targets for promoting tumour immunity and implications for checkpoint blockade. Leukemia 2016;30:1716–1724. [DOI] [PubMed] [Google Scholar]
- 6.Plaumann J, Engelhardt M, Awwad MHS, et al. . IL-10 inducible CD8(+) regulatory T-cells are enriched in patients with multiple myeloma and impact the generation of antigen-specific T-cells. Cancer Immunol Immunother 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Muthu Raja KR, Kubiczkova L, Rihova L, et al. . Functionally suppressive CD8 T regulatory cells are increased in patients with multiple myeloma: a cause for immune impairment. PLoS One 2012;7:e49446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tang M, Diao J, Gu H, Khatri I, Zhao J, Cattral MS. Toll-like Receptor 2 Activation Promotes Tumor Dendritic Cell Dysfunction by Regulating IL-6 and IL-10 Receptor Signaling. Cell Rep 2015;13:2851–2864. [DOI] [PubMed] [Google Scholar]
- 9.Hope C, Foulcer S, Jagodinsky J, et al. . Immunoregulatory roles of versican proteolysis in the myeloma microenvironment. Blood 2016;128:680–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Arana P, Zabaleta A, Lasa M, et al. . High-Throughput Characterization and New Insight into the Role of Tumor Associated Macrophages (TAMs) in Multiple Myeloma (MM). Blood 2016;128:482–482. [Google Scholar]
- 11.Hope C, Emmerich PB, Papadas A, et al. . Versican-Derived Matrikines Regulate Batf3-Dendritic Cell Differentiation and Promote T Cell Infiltration in Colorectal Cancer. J Immunol 2017;199:1933–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Spranger S, Dai D, Horton B, Gajewski TF. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017;31:711–723 e714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nandadasa S, Foulcer S, Apte SS. The multiple, complex roles of versican and its proteolytic turnover by ADAMTS proteases during embryogenesis. Matrix Biol 2014;35:34–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Rosenblatt J, Avivi I, Vasir B, et al. . Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res 2013;19:3640–3648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jensen JL, Rakhmilevich A, Heninger E, et al. . Tumoricidal Effects of Macrophage-Activating Immunotherapy in a Murine Model of Relapsed/Refractory Multiple Myeloma. Cancer Immunol Res 2015;3:881–890. [DOI] [PMC free article] [PubMed] [Google Scholar]