46
1 A circulating T-cell differentiation marker to predict response to immune 1 checkpoint inhibitors 2 3 Takayoshi Yamauchi 1 , Toshifumi Hoki 1 , Takaaki Oba 1 , Vaibhav Jain 1 , Hongbin Chen 2 , 4 Kristopher Attwood 4 , Sebastiano Battaglia 1,5 , Saby George 2,3 , Gurkamal Chatta 2 , Igor Puzanov 2 , 5 Carl Morrison 6 , Kunle Odunsi 1,7,8 , Brahm H. Segal 2,3,8 , Grace K. Dy 2 , Marc S. Ernstoff 2,3 , 6 Fumito Ito 1,8,9,10 * 7 8 1 Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 9 2 Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York 10 3 Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical 11 Sciences, the State University of New York, Buffalo, NY 12 4 Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 13 5 Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, 14 Buffalo, NY 15 6 Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 16 7 Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, 17 NY 18 8 Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 19 9 Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 20 10 Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical 21 Sciences, the State University of New York, Buffalo, NY 22 23 . CC-BY-NC-ND 4.0 International license available under a (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made The copyright holder for this preprint this version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844 doi: bioRxiv preprint

A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

1

A circulating T-cell differentiation marker to predict response to immune 1

checkpoint inhibitors 2

3

Takayoshi Yamauchi1, Toshifumi Hoki1, Takaaki Oba1, Vaibhav Jain1, Hongbin Chen2, 4

Kristopher Attwood4, Sebastiano Battaglia1,5, Saby George2,3, Gurkamal Chatta2, Igor Puzanov2, 5

Carl Morrison6, Kunle Odunsi1,7,8, Brahm H. Segal2,3,8, Grace K. Dy2, Marc S. Ernstoff2,3, 6

Fumito Ito1,8,9,10 * 7

8

1 Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 9

2 Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York 10

3 Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical 11

Sciences, the State University of New York, Buffalo, NY 12

4 Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 13

5 Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, 14

Buffalo, NY 15

6 Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 16

7 Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, 17

NY 18

8 Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 19

9 Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 20

10 Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical 21

Sciences, the State University of New York, Buffalo, NY 22

23

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 2: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

2

Please address all correspondence to F. Ito 24

Elm and Carlton Streets, Buffalo, NY 14263 25

Phone: 716-845-1300 FAX: 716-845-1595 26

Email: [email protected] 27

28

Running title: CX3CR1 as a dynamic blood-based T-cell predictive biomarker 29

30

Grant support: This work was supported by National Cancer Institute (NCI) grant 31

P30CA016056 involving the use of Roswell Park Comprehensive Cancer Center’s Flow 32

Cytometry Core, Clinical Research and Pathology Network, Biostatistics & Statistical Genomics 33

Shared Resource, and Genomic Shared Resources, and by the National Center for Advancing 34

Translational Sciences of the NIH (UL1TR001412) to the University of Buffalo. This work was 35

also supported by grants from the Roswell Park Alliance Foundation, the Melanoma Research 36

Alliance, Department of Defense Lung Cancer Research Program (LC180245), and the NIH/NCI 37

(K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 38

U01CA154967, P50CA159981-01A1 (K. Odunsi), Uehara Memorial Foundation (T. Oba), and 39

Astellas Foundation for Research on Metabolic Disorders and the Nakatomi Foundation (T. 40

Yamauchi). 41

42

Competing interests: Igor Puzanov: Amgen consultant. Marc S. Ernstoff receives clinical trial 43

support from Merck, Bristol Myers Squibb, Alkermes, EMD Serono and serves on a BMS 44

DMSC and consultant for ImmuNext, Alkermes, EMD Serono, Merck and BMS. Kunle Odunsi 45

co-founder of Tactiva Therapeutics and receives research support from Astra Zeneca and Tessaro. 46

47

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 3: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

3

Abstract 48

Immune checkpoint inhibitors (ICI) have revolutionized treatment for various cancers; however, 49

durable response is limited to only a subset of patients. Discovery of blood-based biomarkers 50

that reflect dynamic change of the tumor microenvironment, and predict response to ICI will 51

markedly improve current treatment regimens. Here, we investigated a role of CX3C chemokine 52

receptor 1 (CX3CR1), a marker of T-cell differentiation, in predicting response to ICI therapy. 53

Successful treatment of tumor-bearing mice with ICI increased the frequency and T-cell receptor 54

clonality of the peripheral CX3CR1+CD8+ T-cell subset that included an enriched repertoire of 55

tumor-specific and tumor-infiltrating CD8+ T cells. Furthermore, an increase in the frequency of 56

the CX3CR1+ subset in circulating CD8+ T cells early after initiation of anti-PD-1 therapy 57

correlated with response and survival in patients with non-small cell lung cancer (NSCLC). 58

Taken together, these data support T-cell CX3CR1 expression as a blood-based dynamic 59

biomarker to predict response to ICI therapy. 60

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 4: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

4

Cancer immunotherapies that target the immune checkpoints, such as cytotoxic T 61

lymphocyte associated antigen 4 (CTLA-4), programmed cell death protein-1 (PD-1), and PD1 62

ligand-1 (PD-L1), have transformed the therapeutic landscape of a variety of malignancies1-3. 63

However, despite unprecedented and durable clinical responses seen across diverse tumor types, 64

only a fraction of patients achieve durable responses. Moreover, unusual response patterns such 65

as pseudoprogression and delayed response, a dichotomous outcome, potentially severe toxicity, 66

and high cost indicate a critical need for a reliable predictive biomarker4-7. 67

Baseline PD-L1 expression on immune and tumor cells, preexisting infiltrating CD8+ T 68

cells, and tumor mutational burden (TMB) correlate with response2-11; however, the use of these 69

pretreatment markers are hampered by the significant overlap between responders and non-70

responders, limited quantity and quality of the tissue, and/or lack of standardization12-14. Analysis 71

of serially collected tumor samples could aid in the assessment of evolution of the tumor 72

microenvironment (TME) during immune checkpoint inhibitor (ICI) therapy15-18; however, this 73

approach is invasive and challenging for visceral tumors such as non-small cell lung cancer 74

(NSCLC). Discovery of dynamic circulating immune biomarkers that reflect the evolution of 75

adaptive immunity in the TME and are early predictors of clinical response to ICI would be of 76

value to guide selection of patients most likely to benefit from ICI therapy. 77

Emerging blood-based biomarkers such as exosomal PD-L1, TMB and T cell receptor 78

(TCR) sequence in cell-free DNA, and hypermutated circulating tumor DNA associate with 79

response19-23; however, these approaches require complex platforms, and/or bioinformatics 80

analysis, that limit their widespread application in community-based clinical practice. Since ICI 81

targets T-cell regulatory pathways, utility of surface and intracellular proteins expressed on T 82

cells have been investigated as a potential biomarker for response8, 9, 24-28. Of these, the 83

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 5: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

5

proliferation marker Ki-67 has been extensively investigated. However, most studies showed Ki-84

67 expression only transiently increased in subsets of peripheral blood (PB) CD8+ T cells after 85

the first cycle with unclear predictive and prognostic value as a stand-alone predictive biomarker 86

for ICI8, 9, 24-27. 87

Recently, CX3C chemokine receptor 1 (CX3CR1) was found to be a marker of T-cell 88

differentiation, where CX3CR1+CD8+ T cells were progeny of CX3CR1-CD8+ T cells, and 89

exhibited robust cytotoxicity in anti-viral immunity29, 30. Mechanistically, CX3CR1 is stably 90

expressed on CD8+ T cells through unidirectional differentiation from CX3CR1-CD8+ T cells 91

during the effector phase30-32, which theoretically provides an advantage as a biomarker 92

compared with transiently expressed molecules on T cells. Indeed, an increased frequency of 93

PB CX3CR1+CD8+ T cells has been observed in a few patients who responded to anti-VEGF 94

and anti-PD-L1 antibodies (Ab)33 or chemotherapy and anti-PD-1 Ab34; however, no studies 95

have evaluated the utility of CX3CR1 on T cells as a blood-based predictive biomarker only 96

for ICI therapy. 97

Here, we hypothesized that changes in the frequency of PB CX3CR1+CD8+ T cells 98

would correlate with response to ICI and help identify responders versus non-responders early 99

after initiation of therapy. We investigated the frequency of CX3CR1+CD8+ T cells in PB 100

before and during ICI therapy, and delineated the TCR repertoire in peripheral 101

CX3CR1+CD8+ T-cell subsets and CD8+ tumor-infiltrating lymphocytes (TILs) using 102

preclinical models. To understand the clinical utility of CX3CR1 as a circulating T-cell 103

biomarker, we analyzed longitudinal PB samples from patients with NSCLC undergoing anti-104

PD-1 therapy and evaluated the predictive and prognostic value of changes in the frequency of 105

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 6: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

6

PB CX3CR1+ CD8+ T cells. Our results support circulating CX3CR1+CD8+ T cells as an early 106

on-treatment biomarker for clinical response to anti-PD-1 therapy. 107

108

Results 109

Increased frequency of PB CX3CR1+CD8+ T cells after effective ICI therapy 110

While ICI are known to restore cytokine production and proliferation of effector T 111

cells35, whether or not ICI affect differentiation of tumor-specific T cells remains elusive. To 112

this end, we evaluated CX3CR1 expression, a marker of T-cell differentiation in combination 113

with CD27 (refs.29-31), on PB CD8+ T cells before and during treatment with anti-PD-L1 and 114

anti-CTLA-4 Ab or isotype Ab (NT: no-treatment) in two mouse tumor models, MC38 and 115

CT26 colon adenocarcinoma (Fig. 1A). Significant tumor growth delays (Fig. 1B) and 116

increased frequency of CX3CR1+CD8+ T cells (Fig. 1C) were observed in MC38 and CT26 117

tumor-bearing mice treated with ICI compared to mice receiving isotype Ab. Next, we used a 118

tetramer (Tet) to detect CD8+ T cells specific for mutated Adpgk protein (AdpgkMut) in MC38 119

and shared tumor-associated antigen (TAA), gp70 in CT26 tumors36, 37, and found 120

substantially increased frequency of CX3CR1+Tet+CD8+ T cells in both tumor models (Fig. 121

1D), suggesting that T-cell differentiation after ICI therapy occurs in tumor-specific CD8+ T 122

cells. 123

124

Changes in the frequency of PB CX3CR1+CD8+ T cells associate with response to ICI 125

Although both anti-PD-L1 Ab and anti-CTLA-4 Ab target subsets of exhausted-like 126

CD8+ T cells, they do so through distinct cellular mechanisms38. Therefore, we evaluated 127

CX3CR1 expression of PB CD8+ T cells in CT26-bearing mice treated with either anti-PD-L1 128

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 7: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

7

Ab, anti-CTLA-4 Ab or both. Increased frequency as well as the percentage change of the PB 129

CX3CR1+CD8+ T cells in individual mice were seen after either monotherapy or combined 130

ICI therapy compared to no treatment (Fig. 1E). We also examined whether the frequency of 131

the CX3CR1+ subset in Tet+CD8+ T cells correlates with treatment response. To this end, we 132

drew PB and measured CT26 tumor size in individual mice before and 1 week after treatment 133

with either anti-PD-L1 Ab, anti-CTLA-4 Ab or both. We found that frequency of the 134

CX3CR1+ subset in PB Tet+CD8+ T cells correlated with response to ICI, suggesting the 135

potential utility of CX3CR1 as a blood-based T-cell biomarker to predict response to ICI (Fig. 136

1F). 137

138

CX3CR1 but not Ki-67 is stably upregulated in PB CD8+ T cells during ICI therapy 139

To gain insight into the PB CX3CR1+ CD8+ T cells, we evaluated the expression of the 140

nuclear protein Ki-67, a marker for proliferation that is upregulated in subsets of PB CD8+ T 141

cells in response to ICI8, 9, 24-27. The levels of Ki-67 expression in the CX3CR1+CD8+ T cells 142

were significantly higher than in the CX3CR1- (CD27loCX3CR1- and CD27hiCX3CR1-) 143

subsets 2 weeks after ICI therapy in CT26 tumor-bearing mice (Fig. 2A). We next examined 144

whether an increased expression of CX3CR1 on PB CD8+ T cells is transient or sustained 145

during ICI therapy. Increased frequency of CX3CR1+ subset was seen in both CD8+ and 146

Tet+CD8+ T cells starting from day 7, which remained high during treatment (Fig. 2B). In 147

contrast, Ki-67 expression peaked at day 14, and returned to the baseline at day 21 (Fig. 2B). 148

149

Tumor-specific CD8+ T cells are enriched in the CX3CR1+ subset in PB 150

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 8: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

8

Identification of circulating T-cell biomarkers that enrich tumor-reactive T cells may 151

facilitate discovery of dynamic predictive marker of response to ICI. First, we assessed the 152

frequency of Tet+CD8+ T cells within the PB CX3CR1+ and CX3CR1- subsets in MC38 and 153

CT26 tumor-bearing mice (Supplementary Fig. 1A). We found more Tet+CD8+ T cells in the 154

CX3CR1+ subset than in the CX3CR1- subsets even before treatment although the frequency 155

varied between individual mice (Supplementary Fig. 1B). Next, we evaluated change in the 156

frequency of Tet+CD8+ T cells during ICI therapy in CT26 tumor-bearing mice. The frequency 157

of Tet+CD8+ T cells remained higher in the CX3CR1+ subset than in the CX3CR1- subsets, and 158

became less variable between individual mice at day 21 (Fig. 2C). 159

160

Clonally expanded TCR repertoires of CD8+ TILs are enriched in the peripheral CX3CR1+ 161

subset during ICI therapy. 162

The high frequency of tumor-specific CD8+ T cells in the CX3CR1+ subset before and 163

during ICI treatment is suggestive that heterogeneous tumor-infiltrating CD8+ T cells are also 164

enriched in the CX3CR1+ subset. To this end, we performed TCR sequencing on isolated 165

CD8+ TILs and splenic CD27loCX3CR1-, CD27hiCX3CR1-, and CX3CR1+CD8+ T cells from 166

MC38 tumor-bearing mice treated with combined ICI therapy (Supplementary Fig. S2A, B). 167

Comparison of the TCR repertoire in CD8+ TILs and three subsets of splenic CD8+ T cells 168

demonstrated high degree of overlap in TCR usage between CD8+ TILs and splenic 169

CX3CR1+CD8+ T cells (Fig. 3A; Supplementary Fig. S3) as determined by the Morisita’s 170

overlap index39. 171

Next, we evaluated TCR clonality in CD8+ TILs and three subsets of splenic CD8+ T 172

cells 2 weeks after ICI therapy (Fig. 3B). The 100 most abundant TCR clones (colored) 173

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 9: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

9

comprised more than 50% of TCR sequences in the splenic CX3CR1+ subset and CD8+ TILs 174

while the majority of TCR sequences in the splenic CX3CR1- subsets were constituted of less 175

frequent clones (purple). To quantify the skewness of the clonal distribution, we measured the 176

Gini index, and found similarly higher clonality in the splenic CX3CR1+ subset and CD8+ 177

TILs compared to splenic CX3CR1- subsets. Lorenz curves for splenic CX3CR1+ subset and 178

CD8+ TILs were far from the equidistribution line, suggesting unequal distribution and 179

skewing of the TCR repertoire. 180

181

ICI therapy induces high degree of TCR sequence similarity and clonality between CD8+ 182

TILs and the peripheral CX3CR1+ subset. 183

TCRs that recognize the same antigen may not be the exact TCR clonotypes but have 184

highly homologous sequences and share similar sequence features40, 41. Unlike Morisita’s 185

overlap index, a bioinformatics program, ImmunoMap42, allows us to analyze biological 186

sequence similarity in between peripheral and intratumoral CD8+ T cells. Compared to the 187

CX3CR1- subsets, splenic CX3CR1+CD8+ T-cell subsets contain highly frequent clonally 188

expanded CD8+ T cells indicated by the large circles in the end of many different branches of 189

the tree 2 weeks after ICI therapy (Fig. 3C; Supplementary Fig. S4A, B). 190

Analysis of the top six dominant CDR3β amino acid (AA) sequences in splenic 191

CX3CR1+CD8+ T cells and CD8+ TILs revealed they shared a highly frequent AA sequence 192

(CASSLVGNQDTQYF) in all three independent experiments (Table 1, yellow highlighted). 193

Although the most frequent AA sequence, CASSPRLGDNYAEQFF, in splenic 194

CX3CR1+CD8+ T cells was not identified in CD8+ TILs in the same experiment (Exp. #1 in 195

Table 1A), this AA sequence had a high degree of sequence homology with dominant AA 196

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 10: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

10

sequences, CASSPGYAEQFF and CASSPGQGYAEQFF in CD8+ TILs, located in the same 197

branch of the dendrogram (Table 1B, blue highlighted; Supplementary Fig. S4A). Similarly, 198

abundant AA sequences, CASSPGRGYEQYF in splenic CX3CR1+CD8+ T cells and 199

CASSSGTYEQYF in CD8+ TILs clustered largely in the same branch, indicating that they 200

shared a high degree of similarity (Exp. #2 in Table 1, green highlighted; Supplementary 201

Fig. S4B). 202

Furthermore, CD8+ TILs and splenic CX3CR1+CD8+ T cells had a similar number of 203

structural motifs that dominated the response, and higher numbers of responding clones that 204

expanded 10 times more than the summation of all other homologous clones in a sample (Fig. 205

3D, E). The TCR diversity score and the Shannon entropy calculations revealed that both 206

populations have similar TCR diversity (Fig. 3F, G). Collectively, these findings suggest 207

TCR repertoires in peripheral CX3CR1+CD8+ T-cell clones reflect the TCR repertoires in 208

CD8+ TILs, and CX3CR1 on PB CD8+ T cells may act as a dynamic biomarker during the 209

course of effective ICI therapy. 210

211

Expansion of the CX3CR1+ subset in PB CD8+ T cells correlates with improved response to 212

anti-PD-1 therapy and survival in patients with NSCLC. 213

We next explored whether changes in the frequency of the CX3CR1+ subset in PB 214

CD8+ T cells correlate with response to ICI in patients. We analyzed PBMC samples from the 215

PB of a cohort of 36 NSCLC patients treated with anti-PD-1 Ab (Pembrolizumab or 216

Nivolumab) (Fig. 4A). PB was obtained before treatment and every 2-6 weeks during therapy 217

for 12 weeks. All patients had pretreatment tumor tissue available to assess PD-L1 expression. 218

The frequency of TILs and TMB were also analyzed as described before43. Baseline 219

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 11: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

11

characteristics of 36 NSCLC patients are described in Supplementary Table S1. Clinical 220

response in individual patients was derived from investigator-reported data per iRECIST 221

criteria44 at the 12 week time point. Overall response rates (ORR) which include complete 222

response (CR) and partial response (PR) were 36.7% and 20% for patients with a PD-L1 223

tumor proportion score (TPS) of 50% or greater and 1-49%, respectively, in line with previous 224

studies45, 46. 225

We evaluated the maximal percent change of the CX3CR1+ subset in PB CD8+ T cells 226

at 3, 6, 9 and 12-weeks relative to baseline. The maximal percent change was substantially 227

higher in responders than non-responders as early as 3 weeks from the initiation of the treatment 228

(Fig. 4B). Next, we obtained estimates of the area under the curve (AUC) and corresponding 229

95% confidence interval (CI) using a logistic regression model, and the optimal cut-off score for 230

discriminating between groups using the Youden’s index criterion47. These analyses revealed 231

that an increase of CX3CR1+CD8+ T-cell subsets by 15.5~21.2% from baseline segregated 232

responders from non-responders at 6-12 weeks (Supplementary Table S2). Hereafter the 233

percent change of the CX3CR1+ subset in PB CD8+ T cells from baseline are designated as a 234

“CX3CR1 score”. Fig. 4C shows longitudinal CD8 T-cell responses in individual patients. 235

We found at least 20% increase of the CX3CR1 score in 92.3% (12/13) of responders 236

compared to only 13.0% (3/23) of non-responders. 237

Based on these results, we hypothesized that a cut-off of at least 20% increase of the 238

CX3CR1 score would correlate with response to anti-PD-1 therapy and assessed the 239

association between the CX3CR1 score and objective response. The maximal CX3CR1 score of 240

at least 20 was strongly associated with ORR (P<0.0001) (odds ratio, 80.00; 95% CI, 7.45 to 241

858.94), while PD-L1 TPS at ≥50% was also correlated with ORR (P=0.005) (odds ratio, 242

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 12: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

12

2.53; 95% CI, 0.25 to 25.39) (Fig. 4D). Next, we analyzed the corresponding sensitivity, 243

specificity, positive predictive value (PPV), and negative predictive value (NPV) of the CX3CR1 244

score and the PD-L1 TPS. The CX3CR1 score demonstrated remarkably high PPV, NPV, 245

sensitivity, and specificity, and identified response in 32/36 (88.9%) while a PD-L1 TPS of at 246

least 50% had suboptimal PPV and specificity, and correctly identified response only in 16/36 247

(44.4%) (Fig. 4E). Notably, tumor tissues were available for assessing frequency of TILs and 248

TMB for only 66.6% (24/36) and 61.1% (22/36) of NSCLC patients, respectively 249

(Supplementary Table S3), suggesting limitation of these analyses, in line with previous 250

reports48, 49. Lastly, we evaluated correlation between the CX3CR1 score and survival. We 251

found that at least 20% increase of the CX3CR1 score was associated with better progression 252

free (PFS) and overall survival (OS) (Fig. 4F). Median PFS and OS among patients with a 253

CX3CR1 score of less than 20% were 5.7 months (95% CI, 2.2 to 11.8), and 8.6 months (95% 254

CI, 1.4 to 9.4), respectively, while median PFS and OS among patients with a CX3CR1 score 255

of at least 20% were not reached. Taken together, the CX3CR1 score provides highly accurate 256

prediction of a patient’s clinical response early on-treatment and correlates with survival. 257

258

Discussion 259

The lack of robust predictive biomarker for response is a major obstacle of ICI therapy. 260

Blood-based mechanism-driven dynamic biomarkers that reflect constantly evolving TME would 261

be ideal, and intense efforts are ongoing to identify circulating biomarkers for ICI response4-7, 19-262

23. Here, we provide evidence in tumor-bearing mice that: 1) effective ICI therapy correlates with 263

the increased frequency and TCR clonality of peripheral CX3CR1+CD8+ T cells that identify an 264

enriched repertoire of neoantigen- and TAA-specific CD8+ T cells; 2) the frequency of 265

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 13: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

13

CX3CR1+ but not Ki-67+ PB CD8+ T cells remained elevated during ICI therapy; and 3) there 266

are high degree of TCR sequence overlap and similarity between CD8+ TILs and the peripheral 267

CX3CR1+ subset during ICI therapy. Furthermore, analysis of longitudinal PB samples obtained 268

from a cohort of NSCLC patients highlights the potential clinical utility of CX3CR1 as a useful 269

blood-based biomarker to predict response to ICI early after initiation of therapy. 270

Mechanistically, there are some potential advantages for CX3CR1 as a blood-based 271

biomarker. First, unlike other molecules such as Ki-67 and PD-1 which are transiently 272

upregulated on T cells after activation, CX3CR1 is stably expressed on virus- and tumor-specific 273

CD8+ T cells upon differentiation30-32. In agreement with this, we found CX3CR1 but not Ki-67 274

remains elevated in tumor-specific CD8+ T cells during ICI therapy in preclinical models. 275

Additionally, many responding NSCLC patients in our cohort maintained the level of CX3CR1 276

expression on CD8+ T cells above their baseline. Second, given the low levels of CXCR3 277

expression in CX3CR1hi CD8+ T cells30, which is required to traffic to tumors50, it is possible that 278

CX3CR1+CD8+ T cells remain in circulation, and might not actively traffic to the tumor unless 279

fractalkine (CX3CL1), the ligand of CX3CR1 is produced from the TME. In support with this 280

notion, a recent study showed a higher fraction of CX3CR1+CD8+ T cells in PB compared with 281

tumors in NSCLC patients51. Furthermore, we have recently reported that adoptively transferred 282

tumor-specific CX3CR1-CD8+ T cells generate CX3CR1+CD8+ T cells upon in vivo stimulation, 283

traffic to the TME, and mediate effective regression of established tumors32. In contrast, 284

CX3CR1+CD8+ T cells had no impact on established tumors compared with the CX3CR1- subset 285

and became dominant in PB in a preclinical model32. These features of CX3CR1+CD8+ T cells 286

might have contributed to the greater accuracy of the CX3CR1 score in our cohort and make 287

them uniquely suitable for a circulating T-cell biomarker. 288

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 14: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

14

Increased TCR clonality can be identified in tumors and associates with response to 289

anti-PD-1 therapy10; however, the role of PB TCR clonality in predicting response to ICI 290

remains elusive. PB T cells contain highly diverse TCR repertoires, the majority of which are 291

not specific to the tumor; therefore, changes of PB TCR clonality might be difficult to detect 292

even in responders. Identification of markers to detect tumor-specific T cells in PB might 293

overcome this limitation. PD-1 was found to be a cell-surface marker to identify PB neoantigen-294

reactive T cells52, and a recent study showed TCR clonality changes can be observed in PB 295

PD-1+CD8+ T cells53. Although additional clinical studies are required to determine the 296

predictive and prognostic significance of the TCR clonality in PB CX3CR1+CD8+ T cells, our 297

findings also provide insight into the utility of TCR clonality in the peripheral T-cell subset. Our 298

findings of high clonality in peripheral CX3CR1+CD8+ T cells that contain intratumoral CD8+ 299

T-cell repertoires also align with emerging evidence from high-dimensional profiling of PB T 300

cells by single-cell RNA and/or TCR sequencing showing expansion of cytotoxic effector 301

memory CD8+ T cells that include novel intratumoral clones in patients responding to ICI 302

therapy22, 54, 55. 303

There is an unmet clinical need in establishing reliable predictive biomarkers for 304

combined anti-PD-1 and anti-CTLA-4 therapy, where the risk of severe immune-related adverse 305

events (irAEs) is as high as the proportion of patients responding to combined ICI. High NPV 306

early on-treatment biomarkers could reliably identify a lack of response, and minimize 307

unnecessary treatment, toxicity and associated cost. This is especially relevant for patients with 308

NSCLC who have various treatment options available. Our analyses demonstrated that the 309

CX3CR1 score ≥20 at 6-12 weeks could discriminate responders from non-responders with 310

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 15: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

15

high NPV, suggesting that the CX3CR1 score may help make a decision about whether to 311

continue ICI or switch to alternative more effective treatment. 312

The CX3CR1 score might be convenient to use in clinical setting from technical and 313

analytical perspective. While we isolated PBMCs in this study, the same analysis can be done 314

with as little as 1 ml of whole blood without isolating PBMC by density gradient centrifugation. 315

Unlike intracellular proteins, there is no need for fixation/permeabilization procedure to stain 316

CX3CR1. The CX3CR1+ subset can be easily distinguishable from the CX3CR1- subsets in PB 317

CD8+ T cells unlike other markers such as PD-1, where the boundary between positive and 318

negative populations might be difficult to set56. The CX3CR1 score can be obtained by 319

traditional fluorescence flow cytometric analysis, and does not require NGS platform, complex 320

algorithm, or bioinformatics analysis. Thus, the results can be readily available for prompt 321

feedback to oncologists and patients. 322

Tumor PD-L1 expression predicts the likelihood of response to ICI and is used as a 323

companion diagnostic for NSCLC but not for several other malignancies such as melanoma. 324

Therefore, it remains elusive whether the CX3CR1 score would be useful in patients with 325

other types of cancer. A recent study showed T-cell invigoration to tumor burden ratio was 326

associated with response to anti-PD-1 Ab and clinical outcome in melanoma patients24. 327

Although it was not within the scope of our study, one future area of investigation would be to 328

compare the utility of different PB T-cell biomarkers in the same disease, or evaluate whether 329

combining multiple biomarkers could improve predictive and prognostic values. 330

In summary, our findings demonstrate that CX3CR1 is a useful dynamic circulating T-331

cell biomarker, and that at least 20% increase of the CX3CR1+ subset in PB CD8+ T cells 332

identifies NSCLC patients responding to anti-PD-1 therapy early on-treatment. Our study 333

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 16: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

16

provides a rationale for further investigation to test the utility of the circulating T-cell 334

differentiation marker for a wide variety of malignancies in larger prospective trials. 335

336

Methods 337

Please see the Supplementary Data. 338

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 17: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

17

Acknowledgments 339

We thank the patients, their families and the clinical study staff, Ms. Elongia Farrell, Noelle 340

Brunsing, Suzanne M. Stack, Rushka Kallicharan-Smith, and Ann Marie DiRaddo (Roswell 341

Park) for their contribution to the study. We acknowledge the NIH Tetramer Core Facility 342

(contract HHSN272201300006C) for provision of MHC-I tetramers, and Dr. Weiping Zou 343

(University of Michigan, Ann Arbor, Michigan) for MC38 cells, Drs. James J. Moon (University 344

of Michigan, Ann Arbor, Michigan), Kenichi Makino and Toshihiro Yokoi (Roswell Park) for 345

technical assistance, and Ms. Sue Hess and Judith Epstein (Roswell Park) for administrative 346

assistance. This work was supported by National Cancer Institute (NCI) grant P30CA016056 347

involving the use of Roswell Park’s Flow and Image Cytometry, Clinical Research and 348

Pathology Network, Biostatistics & Statistical Genomics Shared Resource and Genomic Shared 349

Resources, and by the National Center for Advancing Translational Sciences of the NIH 350

(UL1TR001412) to the University of Buffalo. This work was supported by Roswell Park 351

Alliance Foundation, the Melanoma Research Alliance, Department of Defense Lung Cancer 352

Research Program (LC180245), and NCI grant, K08CA197966 (F. Ito), R01CA188900 (B.H. 353

Segal), R01CA188900 (B.H. Segal), U01CA154967, P50CA159981-01A1 (K. Odunsi), Uehara 354

Memorial Foundation (T. Oba), and Astellas Foundation for Research on Metabolic Disorders 355

and the Nakatomi Foundation (T. Yamauchi). 356

357

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 18: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

18

References 358

1. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. 359 N Engl J Med 363, 711-723 (2010). 360

361 2. Topalian SL, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in 362

cancer. N Engl J Med 366, 2443-2454 (2012). 363 364 3. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced 365

cancer. N Engl J Med 366, 2455-2465 (2012). 366 367 4. Lesterhuis WJ, Bosco A, Millward MJ, Small M, Nowak AK, Lake RA. Dynamic versus 368

static biomarkers in cancer immune checkpoint blockade: unravelling complexity. Nat 369 Rev Drug Discov 16, 264-272 (2017). 370

371 5. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to 372

guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 16, 275-287 373 (2016). 374

375 6. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-checkpoint blockade: 376

response evaluation and biomarker development. Nature reviews Clinical oncology 14, 377 655-668 (2017). 378

379 7. Havel JJ, Chowell D, Chan TA. The evolving landscape of biomarkers for checkpoint 380

inhibitor immunotherapy. Nat Rev Cancer 19, 133-150 (2019). 381 382 8. Herbst RS, et al. Predictive correlates of response to the anti-PD-L1 antibody 383

MPDL3280A in cancer patients. Nature 515, 563-567 (2014). 384 385 9. Powles T, et al. MPDL3280A (anti-PD-L1) treatment leads to clinical activity in 386

metastatic bladder cancer. Nature 515, 558-562 (2014). 387 388 10. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune 389

resistance. Nature 515, 568-571 (2014). 390 391 11. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-392

1 blockade in non-small cell lung cancer. Science 348, 124-128 (2015). 393 394 12. Horn L, et al. Nivolumab Versus Docetaxel in Previously Treated Patients With 395

Advanced Non-Small-Cell Lung Cancer: Two-Year Outcomes From Two Randomized, 396 Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057). J Clin Oncol 35, 397 3924-3933 (2017). 398

399 13. Robert C, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N 400

Engl J Med 372, 320-330 (2015). 401 402

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 19: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

19

14. Taube JM, et al. Association of PD-1, PD-1 ligands, and other features of the tumor 403 immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20, 404 5064-5074 (2014). 405

406 15. Riaz N, et al. Tumor and Microenvironment Evolution during Immunotherapy with 407

Nivolumab. Cell, (2017). 408 409 16. Vilain RE, et al. Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early 410

During Treatment Predict Response to PD-1 Blockade in Melanoma. Clin Cancer Res 23, 411 5024-5033 (2017). 412

413 17. Chen PL, et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields 414

Insight into Biomarkers of Response and Mechanisms of Resistance to Immune 415 Checkpoint Blockade. Cancer Discov 6, 827-837 (2016). 416

417 18. Anagnostou V, et al. Evolution of Neoantigen Landscape during Immune Checkpoint 418

Blockade in Non-Small Cell Lung Cancer. Cancer Discov 7, 264-276 (2017). 419 420 19. Chen G, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with 421

anti-PD-1 response. Nature 560, 382-386 (2018). 422 423 20. Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit 424

in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 24, 1441-1448 425 (2018). 426

427 21. Khagi Y, et al. Hypermutated Circulating Tumor DNA: Correlation with Response to 428

Checkpoint Inhibitor-Based Immunotherapy. Clin Cancer Res 23, 5729-5736 (2017). 429 430 22. Valpione S, et al. Immune-awakening revealed by peripheral T cell dynamics after one 431

cycle of immunotherapy. Nat Cancer 1, 210-221 (2020). 432 433 23. Cescon DW, Bratman SV, Chan SM, Siu LL. Circulating tumor DNA and liquid biopsy 434

in oncology. Nature Cancer 1, 276-290 (2020). 435 436 24. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 437

response. Nature 545, 60-65 (2017). 438 439 25. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-440

targeted therapy in lung cancer patients. Proc Natl Acad Sci U S A 114, 4993-4998 441 (2017). 442

443 26. Kim KH, et al. The First-week Proliferative Response of Peripheral Blood PD-444

1(+)CD8(+) T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors. Clin 445 Cancer Res 25, 2144-2154 (2019). 446

447

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 20: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

20

27. Huang AC, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes 448 in resectable melanoma. Nat Med 25, 454-461 (2019). 449

450 28. Liakou CI, et al. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells 451

to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci U 452 S A 105, 14987-14992 (2008). 453

454 29. Bottcher JP, et al. Functional classification of memory CD8(+) T cells by CX3CR1 455

expression. Nature communications 6, 8306 (2015). 456 457 30. Gerlach C, et al. The Chemokine Receptor CX3CR1 Defines Three Antigen-Experienced 458

CD8 T Cell Subsets with Distinct Roles in Immune Surveillance and Homeostasis. 459 Immunity 45, 1270-1284 (2016). 460

461 31. Gordon CL, et al. Induction and Maintenance of CX3CR1-Intermediate Peripheral 462

Memory CD8(+) T Cells by Persistent Viruses and Vaccines. Cell Rep 23, 768-782 463 (2018). 464

465 32. Yamauchi T, et al. CX3CR1-CD8+ T cells are critical in antitumor efficacy, but 466

functionally suppressed in the tumor microenvironment. JCI insight 5, e133920 (2020). 467 468 33. Wallin JJ, et al. Atezolizumab in combination with bevacizumab enhances antigen-469

specific T-cell migration in metastatic renal cell carcinoma. Nature communications 7, 470 12624 (2016). 471

472 34. Yan Y, et al. CX3CR1 identifies PD-1 therapy-responsive CD8+ T cells that withstand 473

chemotherapy during cancer chemoimmunotherapy. JCI insight 3, e97828 (2018). 474 475 35. Blank C, et al. PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell 476

receptor (TCR) transgenic CD8+ T cells. Cancer research 64, 1140-1145 (2004). 477 478 36. Yadav M, et al. Predicting immunogenic tumour mutations by combining mass 479

spectrometry and exome sequencing. Nature 515, 572-576 (2014). 480 481 37. Huang AY, et al. The immunodominant major histocompatibility complex class I-482

restricted antigen of a murine colon tumor derives from an endogenous retroviral gene 483 product. Proc Natl Acad Sci U S A 93, 9730-9735 (1996). 484

485 38. Wei SC, et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 486

Checkpoint Blockade. Cell 170, 1120-1133.e1117 (2017). 487 488 39. Morisita M. Measuring of the dispersion of individuals and analysis of the distributional 489

patterns. Mem Fac Sci Kyushu Univ Ser E 2, 5-23 (1959). 490 491 40. Glanville J, et al. Identifying specificity groups in the T cell receptor repertoire. Nature 492

547, 94-98 (2017). 493

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 21: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

21

494 41. Dash P, et al. Quantifiable predictive features define epitope-specific T cell receptor 495

repertoires. Nature 547, 89-93 (2017). 496 497 42. Sidhom JW, Bessell CA, Havel JJ, Kosmides A, Chan TA, Schneck JP. ImmunoMap: A 498

Bioinformatics Tool for T-cell Repertoire Analysis. Cancer immunology research 6, 151-499 162 (2018). 500

501 43. Morrison C, et al. Predicting response to checkpoint inhibitors in melanoma beyond PD-502

L1 and mutational burden. Journal for immunotherapy of cancer 6, 32 (2018). 503 504 44. Seymour L, et al. iRECIST: guidelines for response criteria for use in trials testing 505

immunotherapeutics. Lancet Oncol 18, e143-e152 (2017). 506 507 45. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl 508

J Med 372, 2018-2028 (2015). 509 510 46. Herbst RS, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, 511

advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. 512 Lancet 387, 1540-1550 (2016). 513

514 47. Youden WJ. Index for rating diagnostic tests. Cancer 3, 32-35 (1950). 515 516 48. Lim C, et al. Biomarker testing and time to treatment decision in patients with advanced 517

nonsmall-cell lung cancer. Ann Oncol 26, 1415-1421 (2015). 518 519 49. Green MR, Willey J, Buettner A, Lankford M, Neely DB, Ramalingam SS. Molecular 520

testing prior to first-line therapy in patients with stage IV nonsquamous non-small cell 521 lung cancer (NSCLC): A survey of U.S. medical oncologists. Journal of Clinical 522 Oncology 32, 8097-8097 (2014). 523

524 50. Mikucki ME, et al. Non-redundant requirement for CXCR3 signalling during tumoricidal 525

T-cell trafficking across tumour vascular checkpoints. Nature communications 6, 7458 526 (2015). 527

528 51. Guo X, et al. Global characterization of T cells in non-small-cell lung cancer by single-529

cell sequencing. Nat Med 24, 978-985 (2018). 530 531 52. Gros A, et al. Prospective identification of neoantigen-specific lymphocytes in the 532

peripheral blood of melanoma patients. Nat Med 22, 433-438 (2016). 533 534 53. Han J, et al. TCR Repertoire Diversity of Peripheral PD-1(+)CD8(+) T Cells Predicts 535

Clinical Outcomes after Immunotherapy in Patients with Non-Small Cell Lung Cancer. 536 Cancer immunology research 8, 146-154 (2020). 537

538

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 22: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

22

54. Wu TD, et al. Peripheral T cell expansion predicts tumour infiltration and clinical 539 response. Nature 579, 274-278 (2020). 540

541 55. Fairfax BP, et al. Peripheral CD8(+) T cell characteristics associated with durable 542

responses to immune checkpoint blockade in patients with metastatic melanoma. Nat 543 Med 26, 193-199 (2020). 544

545 56. Gros A, et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire 546

infiltrating human tumors. J Clin Invest 124, 2246-2259 (2014). 547 548 549

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 23: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

0

1000

2000

3000

4000

0 5 10 15 20 25 30

NTICI

Figure 1

C D

CD

27 (P

E-C

y7)

4.5 34.8

40.0 78.1

0

20

40

60

% C

X3C

R1+

in C

D8+

Tet+

T ce

lls

NT ICI

P<0.0001

NT ICI

P=0.0091

0

80 100

NT ICI NT ICI

0200400600800

1000

0 5 10 15 20 25

NTICI

Days after tumor injection

Tum

or v

olum

e (m

m3 )

MC38 CT26

Tumors s.c.

Day -21~14 Day 0

Anti-PD-L1 Ab and Anti-CTLA-4 Ab or isotype control Ab (NT: no treatment)

Tumor volume Harvest blood 9 CD27, CX3CR1 in CD8+ and tetramer+ T cells

A B

**

Days after tumor injection

0

5

10

15 %

CX3

CR

1+ in

CD

8+ T

cells

P=0.0002 1.4 10.3

2.1 6.8

CX3CR1 (APC) NT ICI 0

10

20 P=0.0003 NT ICI

MC38

CT26

**

CD

27 (P

E-C

y7)

CX3CR1 (APC)

E F Tu

mor

vol

ume

(fold

cha

nge)

0

5

10

15

10 20 30 40 50 % CX3CR1+ subsets in CD8+ Tet+ T cells

R = -0.541 p = 0.025

0

10

20

% C

X3C

R1+

in

CD

8+ T

cel

ls

chan

ge o

f the

%

CX

3CR

1+ s

ubse

t

P=0.0013 P=0.0007 P=0.0016

P=0.008

P=0.0016 P=0.036

15 **

*** ** Mann-Whitney

Anti-CTLA-4 Ab Anti-PD-L1 Ab

- - - +

+ - + +

0

-10

10

5

-5

P < 0.008 P < 0.008

*** ***

*** **

- - - +

+ - + +

** *

**

60 40 20

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 24: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Figure 1. Changes in the frequency of PB CX3CR1+ CD8+ T cells associates with

response to ICI therapy.

A, Experimental scheme of treatment with immune checkpoint inhibitors (ICI). Anti-PD-

L1 antibody (Ab) and anti-CTLA-4 Ab were administered intraperitoneally every 3 days

and every other day, respectively.

B, Tumor growth curves in MC38 (left) and CT26 (right) tumor-bearing mice treated

with isotype Ab (NT) or ICI (n = 5 mice per group).

C, D, Representative bivariate plot showing CD27 and CX3CR1 expression of CD8+ T

cells (C) and tetramer (Tet)+ CD8+ T cells (D) in peripheral blood (PB) of MC38 (upper)

and CT26 (lower) tumor-bearing mice in different treatments as indicated; numbers

denote percent CX3CR1+ cells. Right panels show the frequency of CX3CR1+ cells

among CD8+ T cells (C) and Tet+ CD8+ T cells (D). PB was harvested at day 13. n = 2

experiments pooled.

E, Frequency of the CX3CR1+ subset among CD8+ T cells (left) and percent change of

CX3CR1+ T-cell subsets (right) in PB of CT26 tumor-bearing mice treated with isotype

control Ab (black), anti-CTLA4 Ab (green), anti-PD-L1 Ab(blue), or combination of

these two Ab (red). (n = 5 - 7 per each group). PB was harvested at day 7.

F, Scatter plot with correlation curve showing relationship between CT26 tumor

volume (fold change) and the frequency of PB CX3CR1+Tet+CD8+ T cells at day 7 in

CT26 tumor-bearing mice as in (E). Correlation is shown using Pearson correlation

coefficients (R) and significance was determined using Spearman correlation.

(B – E) *, P < 0.05; **, P < 0.01; ***, P < 0.001; Mann-Whitney U-test. Values are

mean ± SEM.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 25: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

0

20000

40000

80000

60000

Figure 2

A B 80.2 65.9

48.0

CD8 (FITC)

Ki6

7 (M

FI)

P=0.0038

P=0.0057

Ki67

(BV4

2)

CD27loCX3CR1–

CD27hiCX3CR1– CX3CR1+

CX3CR1 (APC)

Days in treatment 0 7 14 21

Ki67

(MFI

)

0

20000

40000

80000

60000

Ki67

(MFI

)

0

20000

40000

80000

60000

0

5

10

20

15

CX

3CR

1 (%)

CX

3CR

1 (%)

0

50

100

Ki67+ CX3CR1+

CD8 (FITC) CD8 (FITC)

Ki67

(BV4

21)

Days in treatment 0 7 14 21

CD8+ T cells

Tet+ CD8+ T cells

Ki67

(BV4

21)

C One-way ANOVA RM

CD27loCX3CR1– CD27hiCX3CR1– CX3CR1+

0

10

20

30

40

0

Days in treatment

7 14 21

***

% o

f Tet

+ C

D8+

T c

ells

P<0.0005 P<0.03

P<0.02 P<0.0005

* * ***

** **

CD

27 (P

E-C

y7)

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 26: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Figure 2. Phenotypic analysis of PB CX3CR1+ CD8+ T cells before and

during ICI therapy.

A-C, CT26 tumor-bearing mice were treated with anti-CTLA-4 Ab and anti-PD-L1

Ab. Peripheral blood (PB) was obtained before and during treatment.

A, Ki-67 expression of CD27loCX3CR1- (green), CD27hiCX3CR1- (blue), and

CX3CR1+ (red) CD8+ T cells in PB 2 weeks after ICI therapy. Numbers denote

percent Ki-67+ cells. Data panel shows mean fluorescence intensity (MFI) of Ki-

67+ cells in each subset (n=4 per each group).

B, Box and whiskers plots showing MFI of Ki-67+ (blue) and frequency of the

CX3CR1+ subset (red) in CD8+ T cells (upper) and Tet+CD8+ T cells (lower) at

day 0, 7, 14, and 21 in PB (n = 3 - 4 per each group).

C, Frequency of Tet+CD8+ T cells in the CD27loCX3CR1- (green),

CD27hiCX3CR1- (blue), and CX3CR1+(red) subsets at day 0, 7, 14, and 21 in

PB. n = 2 experiments pooled. (A and C) *, P < 0.05; **, P < 0.01; ***, P <

0.0005; repeated-measures one-way ANOVA with Tukey’s multiple comparisons

test. Values are mean ± SEM.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 27: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

A

0

0.5

1.0

P<0.0001 G

ini I

ndex

Cum

ulat

ive

% o

f rea

ds

Cumulative % of unique sequences

CX3CR1+

CD8+ TILs

0

1.0

1.0

CD27loCX3CR1-

CD27hiCX3CR1-

B

Splenic CD27lo CX3CR1- CD8+ T cells and CD8+ TILs

Splenic CX3CR1+ CD8+ T cells and CD8+ TILs

Splenic CD27hi CX3CR1- CD8+ T cells and CD8+ TILs

0

0.1

0.2

0.3

TCR

ove

rlap

(vs.

CD

8+ T

ILs)

C

P<0.007

10-4 10-3 10-2 10-1 10-4 10-3 10-2 10-1 10-4 10-3 10-2 10-1

10-1

10-2

10-3

10-4

Frequency of TCRβ CDR3 AA sequences

CD

8+ T

ILs

CD27loCX3CR1– CD27hiCX3CR1– CX3CR1+

Freq

uenc

y (%

)

0

50

100

Figure 3

0

5

10

15

20

0

10

20

30

40

50

0

1

2

3

4

NS

NS

** ***

*** NS

* *

NS

** **

NS

# of

mot

ifs

# of

clo

nes

Sco

re

Ent

ropy

*** ***

*** ***

NS

NS

*** **

*** ***

NS NS

0.3

0.4

D E F G

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 28: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Figure 3. Effective ICI therapy induces high degree of TCR sequence similarity and clonality between tumor infiltrating CD8+ T cells and peripheral CX3XR1+ CD8+ T cells.

A-G, MC38 tumor-bearing mice were treated with anti-CTLA-4 Ab and anti-PD-L1 Ab.

Three subsets of splenic CD8+ T cells determined by CD27 and CX3CR1 expression

(CD27loCX3CR1-, CD27hiCX3CR1-, and CX3CR1+), and CD8+ tumor-infiltrating

lymphocytes (TILs) were isolated 2 weeks after treatment for TCR repertoire and

clonality analysis.

A, TCR repertoire overlap by Morisita’s index (left) and pairwise scatter plots of the

frequency of TCRβ CDR3 amino acid (AA) sequences between each subset of splenic

CD8+ T cells and CD8+ TILs (right).

B, TCR clonality analysis of three subsets of splenic CD8+ T cells and CD8+ TILs by top

sequence plot (left), Gini index (center), and Lorenz curve (right). The most abundant

100 AA sequences are colored while other less frequent clones are in purple in top

sequence plot. (A and B) Data were analyzed using the one-way ANOVA test with

Tukey’s multiple comparisons to generate P values.

C, Representative overlapped weighted TCR repertoire dendrograms by ImmunoMap

analysis between three subsets of splenic CD8+ T cells (blue) and CD8+ TILs (red). The

distance of the branch ends represents sequence distance, and the size of circles

denotes frequency of sequence. Data shown are representative of three independent

experiments.

D-G, Number of dominant motifs (D), number of clones contributing response (E), TCR

diversity score (F) and Shannon’s entropy (G) of splenic CD27lo CX3CR1- (green),

CD27hi CX3CR1- (blue), CX3CR1+ (red) CD8+ T cells, and CD8+ TILs (black).

NS, not significant, *, P < 0.05; **, P < 0.005; ***, P < 0.0005 by repeated-measures

one-way ANOVA with Tukey’s multiple comparisons test. Values are mean ± SEM.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 29: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

A

B

F

FSC-A

Months

6 0 12 18 24 30 0

25

50

75

100

OS

(%)

P=0.0306

PD-L1 TPS PB CX3CR1 score

0

40 20

60 80

100

OR

R (%

)

≥50% <50% 0

40 20

60 80

100

≥20% <20%

P=0.005

No Objective Response Objective Response

C

D

0

-80

100

-40

20 40

80

150

0

-20

100

3

20

40

80

150

6 9 12 0 3 6 9 12 0

CX

3CR

1 sc

ore

Responders (CR/PR) Non-responders (SD/PD)

weeks weeks

Months

6 0 12 18 24 30 0

25

50

75

100

PFS

(%) CX3CR1 score

≥ 20% (n=15)

CX3CR1 score < 20% (n=21)

P=0.0009

12/31

19/31

P<0.0001

1/5

4/5 12/15

3/15

20/21

1/21

-10

0

10

20

30

40

50

0 3 6 9 12

CR/PRSD/PD

Max

% c

hang

e of

PB

C

X3C

R1+

CD

8+ T

cel

ls

* *

** **

weeks

PD-L1 TPS ≥50% (n=36)

CX3CR1 score ≥20% (n=36)

PPV 38.7% (12/31) 80.0% (12/15) NPV 80.0% (4/5) 95.2% (20/21)

Sensitivity 92.3% (12/13) 92.3% (12/13) Specificity 17.4% (4/23) 87.0% (20/23) Accuracy 44.4% (16/36) 88.9% (32/36)

E

Figure 4

CX3CR1 score ≥ 20% (n=15)

CX3CR1 score < 20% (n=21)

CD

3 (F

ITC

)

CD8 (BV421) C

D4

(PE)

CD8 (BV421)

CX

3CR

1(A

PC)

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 30: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Figure 4. Expansion of the CX3CR1+ subset in PB CD8+ T cells correlates

with improved response to anti-PD-1 therapy and survival in patients with

NSCLC.

A, Gating strategy for identifying CX3CR1+ CD8+ T cells in peripheral mononuclear

blood cells. Cells were first gated for lymphocytes (SSC-A vs. FSC-A) and for

singlets (FSC-H vs. FSC-A).

B, Maximal % change of CX3CR1+ subset in PB CD8+ T cells in responders and

non-responders of 36 NSCLC patients treated with anti-PD-1 therapy by 12 weeks.

CR/PR: complete and partial response, SD/PD: stable and progressive disease. *

P<0.04, **P<0.0001 by Mann-Whitney U-test. Values are median ± SEM.

C, Percent change of the CX3CR1+ subset in PB CD8+ T cells from baseline

(CX3CR1 score) in responders (left) and non-responders (right).

D, Objective response rate (ORR) for high and low PD-L1 tumor proportion score

(TPS) (left) and PB CX3CR1 score (right). ORR was analyzed by Fisher’s exact

test.

E, Comparison of biomarker performance between PD-L1 TPS and CX3CR1 score

at 12 weeks.

F, Kaplan-Meier progression free survival (PFS) (left) and overall survival (OS)

(right) for high versus low CX3CR1 score. The two-tailed P value was calculated

using the log-rank test.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 31: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Table 1: Six most dominant clones in sorted splenic CX3CR1+ CD8+ T cells (A) and CD8+ TILs (B) in MC38-bearing mice treated with CTLA-4 and PD-L1 blockades (n=10 mice / experiment)

aProductive frequency. Bold font with yellow highlight indicates that the clone was present in splenic CX3CR1+ CD8+ T cells and CD8+ TILs at high frequency (>2%) from all three independent experiments. Green and blue-highlighted clones in sorted splenic CX3CR1+ CD8+ T cells (A) and CD8+ TILs (B) have a high degree of sequence homology, respectively.

Exp. CDR3β region sequence Frequencya (%) Sequence also found in CD8+ TILs (%)

#1 (n=10)

CASSPRLGDNYAEQFF CASSDRGRAEQFF CASSLVGNQDTQYF CASGDAQYNNQAPLF CASSPDKYEQYF CASSQSGFAETLYF

4.26 2.65 2.48 2.23 1.84 1.75

#1 (0.24) #2 (2.19) #3 (0.82) #1 (2.68) #2 (9.68) #3 (3.52) #1 (1.82) #3 (3.01) #3 (0.01)

#2 (n=10)

CASSQWGAGNTLYF CASSPGRGYEQYF CTCSPGTASGNTLYF CASSLVGNQDTQYF CASSGRDRKNERLFF CASGDSNERLFF

17.59 2.92 2.89 2.38 1.87 1.38

#2 (1.44) #2 (0.15) #1 (2.68) #2 (9.68) #3 (3.52) #1 (0.02) #2 (0.75)

#3 (n=10)

CAWRGTGSAETLYF CASSGGRQYF CASSLVGNQDTQYF CASSNRVEQYF CASRGDSYNYAEQFF CASSPGRRSGNTLYF

6.33 3.05 2.27 1.87 1.73 1.47

#3 (0.29) #3 (4.17) #1 (2.68) #2 (9.68) #3 (3.52) #3 (0.005) #3 (0.61) #3 (0.22)

Exp. CDR3β region sequence Frequencya (%) Sequence also found in CD8+ TILs (%)

#1 (n=10)

CASTPRDWGVAEQFF CASSRDLGNTGQLYF CASSLELGGPEQYF CASSPGYAEQFF CASSPGQGYAEQFF CASSLVGNQDTQYF

13.61 7.25 3.84 3.68 3.52 2.68

#2 (2.63) #3 (0.01) #3 (0.01) #3 (1.01) #2 (9.68) #3 (3.52)

#2 (n=10)

CASSLVGNQDTQYF CASRRTTNSDYTF CASSSGTYEQYF CASSLELGGREQYF CASSRDLGNTGQLYF CASHLSTSAETLYF

9.68 4.12 3.56 2.77 2.63 2.44

#1 (2.68) #3 (3.52) #3 (1.86) #1 (0.15) #1 (7.25) #3 (0.01)

#3 (n=10)

CTCSETGNSYEQYF CASSGGRQYF CASSLVGNQDTQYF CASSGGWQYF CASGDAQYNNQAPLF CASSPGQNYAEQFF

9.98 4.17 3.52 3.08 3.01 1.98

#1 (2.68) #2 (9.68) #1 (1.81) #1 (0.07) #2 (0.20)

A

B

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 32: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

1

Supplementary Materials and Methods 1

2

Mice 3

Male and female C57BL/6 mice and female Balb/c mice were purchased from the 4

Jackson Laboratories. All mice were 7 to 12 weeks old at the beginning of each experiment, and 5

were housed in the Unit for Laboratory Animal Medicine at the Roswell Park Comprehensive 6

Cancer Center in compliance with the Institutional Animal Care and Use Committee regulations. 7

8

Cell lines 9

MC38 and CT26 murine colon adenocarcinoma cell lines were gifts from Dr. Weiping 10

Zou (University of Michigan) and Dr. Sharon Evans (Roswell Park Comprehensive Cancer 11

Center), respectively. MC38 and CT26 cells were cultured in RPMI (Gibco) supplemented with 12

10% FBS (Sigma), 1% NEAA (Gibco), 2 mM GlutaMAX-1 (Gibco), 100 U/ml penicillin-13

streptomycin (Gibco), and 55 μM 2-mercaptoethanol (Gibco). Cells were authenticated by 14

morphology, phenotype and growth, and routinely screened for Mycoplasma, and were 15

maintained at 37°C in a humidified 5% CO2 atmosphere. 16

17

In vivo mouse studies 18

Male or female C57BL/6 mice and female Balb/c mice were inoculated with 5-8 u 105 19

MC38 and 5 u 105 CT26, respectively per mouse on the right flank by subcutaneous injection on 20

day 0. When tumor volume reached approximately 50 mm3, 200 µg of anti-PD-L1 Ab (clone 21

10F.9G2, BioXcell) and/or 100 µg of anti-CTLA-4 Ab (clone 9H10, BioXcell) were 22

administered intraperitoneally every 3 days and every other day, respectively. Polyclonal syrian 23

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 33: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

2

hamster IgG (BioXcell) and rat IgG2b, κ (BioXcell) were used as isotype control Abs. Tumor 24

volumes were calculated by determining the length of short (l) and long (L) diameters (volume = 25

l2 x L/2). Experimental end points were reached when tumors exceeded 20 mm in diameter or 26

when mice became moribund and showed signs of lateral recumbency, cachexia, lack of 27

response to noxious stimuli, or observable weight loss. 28

29

Single-cell preparations 30

Blood, spleens and tumors were harvested at day 14-42 post MC38 or CT26 tumor 31

implantation. Spleens were homogenized by forcing the tissue through a cell strainer (70 μm; BD 32

Biosciences). Red blood cells in blood and spleen were lysed using ACK Lysis Buffer (Gibco). 33

Tumors were cut into small pieces of 2-4 mm. Single-cell suspensions were obtained by 34

mechanical dispersion consisting of two 30-min incubations at 37°C, 5% CO2 in 5 ml RPMI 35

1640 (Gibco) and tumor dissociation kit (Miltenyi Biotec) in C Tubes (Miltenyi Biotec) 36

interspersed with three mechanical dispersions on a GentleMACS dissociator (Miltenyi Biotec). 37

The tumor cell suspensions were then filtered through a cell strainer (70 μm; BD Biosciences). 38

39

Flow cytometry and cell sorting 40

Surface staining of leukocytes in murine blood, spleens and tumors was performed in 41

FACS buffer (made in house) using monoclonal antibodies against mouse CD3 (145-2C11), 42

CD90.2 (53-2.1), CD4 (GK1.5), CD8 (53-6.7), CX3CR1 (SA011F11) (all BioLegend), CD27 43

(LG.7F9, eBioscience), CD45 (30-F11, Invitrogen), CD8 (KT15 for tetramer staining, 44

Invitrogen), and PD-1 (J43, BD Biosciences). Live/dead cell discrimination was performed using 45

Live/Dead Fixable Aqua Dead Cell Stain Kit or LIVE/DEAD Fixable Near-IR Dead Cell Stain 46

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 34: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

3

Kit (Invitrogen). Samples were incubated with antibodies for 20 min at RT in the dark. We used 47

the tetramer staining assay with peptide-MHC tetramer tagged with PE (H-2Db-restricted 48

ASMTNMELM for MC38-bearing mice and H-2Ld-restricted SPSYVYHQF for CT26-bearing 49

mice (The NIH Tetramer Core Facility)) to analyze the percentages of tumor antigen-specific 50

CD8+ T cells. For intracellular staining, surface-stained cells were fixed and permeabilized using 51

a Foxp3 fixation/permeabilization kit (eBioscience), then stained with anti-Ki67 (16A8, 52

BioLegend) for 30 min. 53

For TCR sequencing of murine splenic CD8+ T cells, single cell suspensions from 54

freshly isolated splenocytes were stained as above. CD45+ CD3+ CD8+ T cells were gated, and 55

CD27lo CX3CR1-, CD27hi CX3CR1-, and CX3CR1+ CD8+ T cells were sorted using BD Aria 56

Sorter. An EasySep Mouse CD8a Positive Selection Kit II (STEMCELL Technologies) was 57

used to isolate murine CD8+ TILs for TCR sequencing. 58

For phenotypic analysis of PBTCs, fresh or cryopreserved PBMC samples were stained 59

with master mix of antibodies for surface stains including CD3 (UCHT1, BD Biosciences), CD4 60

(SK3, BD Biosciences), CD8 (RPA-T8, eBioscience), CD27 (O323, eBioscience), and, CX3CR1 61

(2A9-1, Biolegend). Samples were acquired using LSR II (BD), LSRFortessa (BD) or SONY 62

sorter and data analyzed with FlowJo software v10.1.5 (TreeStar). 63

64

DNA isolation, TCRβ CDR3 region sequencing and repertoire analysis 65

DNA from flow-isolated murine splenic CD8+ T cells and CD8+ TILs, and PB CD8+ T 66

cells was extracted using (QIAamp DNA Micro Kit (QIAGEN)). DNA was quantified using 67

Qubit dsDNA BR Assay (Invitrogen). Amplification and sequencing of TCRβ CDR3 regions 68

was performed using ImmunoSEQ immune profiling system at the survey level (Adaptive 69

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 35: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

4

Biotechnologies)(1). Sequencing was performed on an Illumina NextSeq system using 150 cycle 70

mid-output kit (Illumina Inc.). Processed data were uploaded to the immunoSEQ Platform 71

(Adaptive Biotechnologies) for preliminary bioinformatics analysis. Processed data were 72

downloaded and frequencies/counts for TCR clonotypes and diversity were examined by 73

nucleotide sequences after non-productive reads were filtered out. 74

TCR beta chain CDR3 variable region sequencing was performed using the ImmunoSEQ 75

assay at the survey level (Adaptive Biotechnologies). T-cell repertoires, comprising all detected 76

CDR3 sequences with annotated V and J gene segment identifications were downloaded directly 77

from the ImmunoSEQ Analyzer from Adaptive biotechnologies. Metrics of the complete TCR 78

repertoire in each sample, including the number of productive rearrangements, productive 79

clonality and clonal frequencies were determined using the ImmunoSEQ Analyzer software and 80

confirmed using the LymphoSeq package (2). All other analyses were performed using the 81

LymphoSeq package and custom scripts in the R statistical software environment. Dissimilarity 82

between sample repertoires was calculated using the Morisita’s Index(3), using the vegan 83

package. Differential clone frequencies between samples were determined using the Fisher’s 84

exact test with multiple test correction (Holm method). For differential analysis, only those 85

clones observed with at least 5 cumulative read counts were considered. TCR clonality was 86

calculated as 1-Pielou’s evenness(4) using the immunoSEQ Analyzer®. Clonality values 87

approaching 1 indicate a very skewed distribution of frequencies, whereas values approaching 0 88

indicate that every rearrangement is present at nearly identical frequency. 89

TCR repertoires were visualized as weighted dendrograms using ImmunoMap(5). Only 90

productive sequences with a frequency > 0.1% in the tumor were considered for analysis. 91

Sequence distances were calculated based on sequence alignments scores using a PAM10 92

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 36: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

5

scoring matrix and gap penalty of 30. Circles are overlaid at the end of the branches 93

corresponding to the CDR3 sequences with diameters proportional to the frequency of the 94

sequences observed in the samples. Shannon’s Entropy, Dominant motifs, singular structural 95

clones, singular clones contributing response, and richness of motifs were identified using 96

ImmunoMap. 97

98

99

Data reporting. 100

No statistical methods were used to predetermine sample size. The experiments were not 101

randomized and the investigators were not blinded to allocation during experiments and outcome 102

assessment. 103

104

Study design, patients and specimen collection. 105

Thirty-six patients with naive or previously treated PD-L1 IHC positive non-small cell 106

lung cancer (NSCLC) adenocarcinoma and squamous cell type, undergoing anti-PD-1 Ab 107

(Pembrolizumab or Nivolumab) (Supplemental Table S2) were consented to the collection and 108

storage of blood samples, the analysis of archived tumor tissue, and the review of their medical 109

records under the protocol (I 188310), in accordance with the Institutional Review Board of 110

Roswell Park Comprehensive Cancer Center. Peripheral blood was obtained in EDTA-111

containing tubes before treatment and before each infusion and every 2-6 weeks for 12 weeks. 112

Peripheral blood mononuclear cells (PBMCs) were isolated using Lymphocyte Separation 113

Medium (Corning) density gradient centrifugation and stored using standard protocols. 114

115

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 37: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

6

Assessment of response. 116

Clinical response to anti-PD-1 therapy was determined as best response based on 117

immune related RECIST (iRECIST)(6) at the 12 week time point, and classified as complete 118

response (CR) and partial response (PR) for responders or stable disease (SD) and progressive 119

disease (PD) for non-responders. Objective responses were confirmed by at least one 120

sequential tumor assessment, and objective response rates were calculated as [(CR + PR) ÷ 121

number of patients] × 100. Fisher’s exact test was used to assess the association between PD -122

L1 expression and objective response. 123

124

Immunohistochemical studies 125

The expression of PD-L1 on the surface of tumor cells and frequency of CD8+ T cells 126

were evaluated as described before (7). Briefly, the expression of PD-L1 on the surface of tumor 127

cells was assessed by means of the Dako Omnis platform (Agilent) with the 28–8 pharmDx 128

antibody and scored by published guidelines (8). Serially sectioned tissue was evaluated for 129

lymphocyte infiltration using the anti-CD8 antibody C8/144B (Agilent) and assigned a 130

qualitative score of non-infiltrated, infiltrated, or excluded. Non-infiltrated referred to a sparse 131

number of CD8+ T-cells that infiltrate nests of neoplastic cells and with less than 5% of the 132

tumor showing an infiltrating pattern. Infiltrated represents frequent CD8+ T-cells that infiltrate 133

nests of neoplastic cells in an overlapping fashion at least focally and in more than 5% of the 134

tumor. Excluded represents restriction of more than 95% of all CD8+ T-cells in a tumor to the 135

periphery or interstitial stromal areas and not actively invading nest or groups of neoplastic cells. 136

137

138

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 38: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

7

Tumor mutational burden profiling 139

Tumor mutational burden (TMB) was evaluated as described before (7). In brief, DNA 140

was extracted from each sample and processed for whole-exon DNAseq. TMB was assessed by 141

targeted capture and sequencing of 409 cancer-related genes and amplicon sequencing of 394 142

immune transcripts, respectively, comprising 1.4 Mb of DNA on samples that met validated 143

quality control (QC) thresholds (7). Somatic mutation calling was conducted using Ion Torrent 144

Suite software’s variant caller plugin. Mutational burden (MuB) cutoff was derived from a 145

reference population whereby the median MuB was determined on regular basis. This value was 146

used as a baseline and a high MuB was defined as 2× this median value, or a value of 10.0. 147

148

Statistics 149

Statistical analysis was performed using t-test or Mann-Whitney U test for comparisons between 150

2 groups, 1-way repeated measures ANOVA with Turkey-adjusted multiple comparisons for 151

comparisons more than 2 groups, or the Mantel-Cox method (log-rank test) for survival analysis 152

using GraphPad Prism 7.03 (GraphPad Software) and the R statistical software. TCR repertoire 153

analysis was performed using ImmunoSEQ software (Adaptive Biotechnologies) and 154

ImmunoMap. P < 0.05 was considered statistically significant. Data are presented as mean ± 155

SEM except for Figure 4B (median ± SEM). 156

157

158

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 39: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

8

References 159

1. Carlson CS, Emerson RO, Sherwood AM, Desmarais C, Chung MW, Parsons JM, et al. 160

Using synthetic templates to design an unbiased multiplex PCR assay. Nature 161

communications 2013;4:2680. 162

2. Coffey D. LymphoSeq: Analyze high-throughput sequencing of T and B cell receptors. . 163

R package version 1140 2019. 164

3. Morisita M. Measuring of the dispersion of individuals and analysis of the distributional 165

patterns. Mem Fac Sci Kyushu Univ Ser E 1959;2(21):5-23. 166

4. Pielou EC. Species-diversity and pattern-diversity in the study of ecological succession. 167

Journal of theoretical biology 1966;10(2):370-83. 168

5. Sidhom JW, Bessell CA, Havel JJ, Kosmides A, Chan TA, Schneck JP. ImmunoMap: A 169

Bioinformatics Tool for T-cell Repertoire Analysis. Cancer immunology research 170

2018;6(2):151-62. 171

6. Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, et al. iRECIST: 172

guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 173

2017;18(3):e143-e52. 174

7. Morrison C, Pabla S, Conroy JM, Nesline MK, Glenn ST, Dressman D, et al. Predicting 175

response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden. 176

Journal for immunotherapy of cancer 2018;6(1):32. 177

8. Dako. PD-L1 IHC 28–8 pharmDx: Non-Squamous Non-Small Cell Lung Cancer 178

[interpretation manual]. Santa Clara, CA: Dako 2017. 179

180

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 40: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

B

A

0

10

5

0

10

20

30

% o

f gp7

0-sp

ecifi

c Te

t+ in

CD

8+ T

cel

ls

% o

f Adp

gkM

ut-s

peci

fic

Tet+

in C

D8+

T c

ells

MC38 CT26

Supplementary Figure 1 Yamauchi et al

P<0.005 * * *

** ** ** P<0.0001

13.3 0.6

0.2

CD8 (FITC)

Tet (

Adpg

k)

CD27loCX3CR1–

CD27hiCX3CR1– CX3CR1+

CX3CR1 (APC)

CD

27 (P

E-C

y7)

Tet (

Adpg

k)

Tet (

Adpg

k)

31.1 2.0

0.1

CD8 (FITC)

Tet (

gp70

)

CD27loCX3CR1–

CD27hiCX3CR1– CX3CR1+

CD

27 (P

E-C

y7)

CX3CR1 (APC)

Tetr

(gp7

0)

Tet (

gp70

)

MC38 CT26

CD8 (FITC) CD8 (FITC) CD8 (FITC) CD8 (FITC)

Supplementary Figure S1. Related to Figure 2. (A) Representative flow cytometric plots

showing the frequency of tetramer (Tet)+ CD8+ T cells in the CD27lo CX3CR1- (green),

CD27hi CX3CR1- (blue), and CX3CR1+ (red) subsets in peripheral blood (PB) of MC38

(left) or CT26 (right) tumor-bearing mice treated with immune checkpoint inhibitors (ICI;

anti-PD-L1 Ab and anti-CTLA-4 Ab) for 14 days; numbers denote percent Tet+ cells. We

used a tetramer to detect CD8+ T cells specific for mutated Adpgk protein (AdpgkMut) in

MC38 and shared tumor-associated antigen (TAA), gp70 in CT26 tumor models

(B) Frequency of Tet+ T cells in 3 subsets of PB CD8+ T cells at baseline in MC38 (left)

and CT26 (right) tumor-bearing mice. n=3 experiments pooled. * P<0.005, **P<0.0001 by

Mann-Whitney U-test. Values are mean ± SEM.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 41: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

CX3CR1 (APC)

CD

27 (P

E-C

y7)

MC38 tumors s.c.

Day -14 Day 0

Anti-PD-L1 Ab + Anti-CTLA-4 Ab

• Harvest spleens and tumors from 10 mice • Sort splenic CD27loCX3CR1-,

CD27hiCX3CR1-, and CX3CR1+ CD8 + T-cell subsets, and CD8 + TILs

CD27loCX3CR1– CD27hiCX3CR1– CX3CR1+

100% 95.8% 99.7%

Day 13

Supplementary Figure 2 Yamauchi et al

Supplementary Figure S2. Related to Figure 3. (A) Experimental scheme

of treatment with immune checkpoint inhibitors (ICI), and isolation of three

subsets of splenic CD8+ T cells and CD8+ TILs for TCR repertoire and

clonality analysis for figure 3, supplementary figures S3 - S5, and

supplementary table S1. (B) Representative flow cytometric plots showing

the frequency of splenic CD27lo CX3CR1- (green), CD27hi CX3CR1- (blue),

and CX3CR1+ (red) CD8+ T cells after flow-sort.

B

A

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 42: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

#2 Sp CX3CR1+

#3 Sp CX3CR1+

#1 CD8+ TILs

#2 CD8+ TILs

#3 CD8+ TILs

#1 Sp CX3CR1+

0.283

0.189

0.167

0.02 0.006

0.001

0.003 0.001 0.007 0.005 0 0.057 0.011 0.016 0.078

0.011

0.017 0.002 0.04 0.002 0.005 0.05 0.006 0.016 0.145 0.02

0.049 0.023 0 0.001 0.061 0.003 0.004 0.041 0.017 0.01

0.001 0 0.002

0.002

0

0.042

0.02

0.037 0.001 0.007 0.001

0.002

0.022

0.003

0.035 0.003 0 0

0.013

0.001

0

0.033 0.005

0.051

0.003 0.004 0.002

0.003 0.001 0.025

#2 Sp CD27hiCX3CR1-

#3 Sp CD27hiCX3CR1-

#1 Sp CD27hiCX3CR1-

#2 Sp CD27loCX3CR1-

#3 Sp CD27loCX3CR1-

#1 Sp CD27loCX3CR1-

1.0

0.2

Morisita’s index

Supplementary Figure 3 Yamauchi et al

Supplementary Figure S3. Related to Figure 3A. Pairwise TCRβ repertoire

overlaps among different samples from three independent experiments (#1 -

#3). Numbers denote TCR repertoire overlap by Morisita’s index.

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 43: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

02:CASSPGRG‒YEQYF 19:CASSAN‒‒‒YEQYF 22:CASSSTGGAYEQYF 24:CAS‒‒‒GG‒YEQYF 25:CASR‒‒‒DSYEQYF 26:CASSS‒‒DSYEQYF 28:CASS‒‒DGAYEQYF 30:CASS‒‒‒GEYEQYF 50:CASSSGLGEYEQYF 53:CASSKTGG-YEQYF

03:CASSS‒‒GTYEQYF 07:CASSKTG‒GYEQYF 16:CASS‒A‒‒NYEQYF 21:CAS‒‒‒GG‒YEQYF 23:CASSQEGDGYEQYF 30:CASSSGLGEYEQYF 38:CASSLGG‒GYEQYF 55:CASSRTG-GYEQYF 64:CASSLAGTGYEQYF 66:CASSPGLGGYEQYF

01:CASSLVG ‒‒NQDTQYF 32:CASSLVPGGAGDTQYF 34:CASSLLP ‒‒NQDTQYF 41:CASSLLG ‒‒NQDTQYF

04:CASSLVG‒NQDTQYF 18:CASSLIG‒NQDTQYF 37:CASSLLG‒NQDTQYF

CD8 TILs

Sp CX3CR1+

CD8+ T cells

CD8 TILs Sp CD27hi CX3CR1-CD8+ T cells

CD8 TILs Sp CD27lo CX3CR1-CD8+ T cells

Splenic CX3CR1+

CD8+ T cells CD8+ TILs

Splenic CX3CR1+ CD8+ T cells

CD8+ TILs

04:CASSPG‒‒‒‒‒‒YAEQFF 05:CASSPGQG‒‒‒YAEQFF 11:CASS‒‒‒LGQNYAEQFF 13:CASS‒‒‒DGGQYAEQFF 22:CASSRDLSGGNYAEQFF 25:CASS‒‒‒MGANYAEQFF 26:CAST‒‒‒PGQNYAEQFF 34:CASG‒‒‒LGGNYAEQFF 35:CASSG‒‒LGVNYAEQFF 50:CASS‒‒‒DWGGYAEQFF

01:CASSPRLGDN‒‒YAEQFF 10:CASRG‒‒‒‒‒‒GSYAEQFF 27:CASSPG‒‒‒‒‒‒‒ YAEQFF 39:CASSRT‒‒‒GGGYAEQFF 43:CASSSRLGVNAGYAEQFF 46:CASSPGQG‒‒‒‒YAEQFF

04:CASSQSG----FAETLYF 27:CASSQVG---GAETLYF 33:CSSSQSGWGVETLYF

02:CASSRDLGNTGQLYF 54:CASHRG-GNTGQLYF 59:CASSL-RSNTGQLYF

CD8 TILs

Sp CX3CR1+

CD8+ T cells

CD8 TILs Sp CD27hi CX3CR1- CD8+ T cells

CD8 TILs Sp CD27lo CX3CR1- CD8+ T cells

Splenic CX3CR1+

CD8+ T cells CD8+ TILs

Splenic CX3CR1+ CD8+ T cells

CD8+ TILs

Supplementary Figure 4 Yamauchi et al

Supplementary Figure S4. Related to Figure 3C and Supplementary Table S1.

Representative overlapped weighted TCR repertoire dendrograms by ImmunoMap analysis

between three subsets of splenic CD8+ T cells (blue) and CD8+ TILs (red) from two

independent experiments (A and B). Dominant motif analysis clusters homologous

sequences and selects for clusters contributing to significant proportion of the response.

Two dominant motifs are shown representing highly represented structural motifs in each

experiment. The numbers denote the ranking of the frequency within the subset. (red = fully

conserved amino acids (AA), green = semi-conserved AA, black = non-conserved AA)

A

B

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 44: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Supplementary Table S1. Demographic and clinical characteristics of lung cancer patients on this study Patient characteristics n = 36 Median age (range) 68 (49-89) Sex, n (%) Male 14 (39%) Female 22 (61%) Race, n (%) Caucasian 34 (94%) African-American 2 (6%) ECOG PS 0/1 33 (92%) History of smoking Never 4 (11%) Former 22 (61%) Current 10 (28%) Histology, n (%) Adenocarcinoma 22 (61%) Squamous cell carcinoma 13 (36%) Non-small cell lung cancer with giant features 1 (3%) Stage at diagnosis, n (%) II-III 9 (25%) IV 27 (75%) Prior lung surgery, n (%) 6 (17%) Prior chemotherapy, n (%) One line 5 (14%) Two lines 2 (6%) Prior targeted therapy, n (%) Osimertinib and Erlotinib (EGFR) 2 (6%) Dabrafenib/Trametinib (BRAF V600E) 1 (3%) Prior radiation, n (%) Thoracic radiation 15 (42%) Bone radiation 3 (8%) Gamma knife stereotactic radiosurgery 7 (19%) Known brain metastases, n (%) 7 (19%) Study drug, n (%) Nivolumab 2 (6%) Pembrolizumab 34 (94%) Best disease response at 12 weeks, n (%) Complete Response (CR) 0 (0%) Partial response (PR) 13 (36%) Stable disease (SD) 14 (39%) Progressive disease (PD) 9 (25%)

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 45: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Supplementary Table S2 Marker Performance, Related to Figure 4B.

Maximal percent change of the CX3CR1+ subset in PB CD8+ T cells at 3, 6, 9 and 12-weeks relative to baseline was evaluated. Cut-points for discriminating between responders and non-responders were obtained using the Youden’s index criterion. Abbreviations: PB, peripheral blood; Max, maximal; PPV, positive predictive value; NPV, negative predictive value.

PB CX3CR1+CD8+ T cells Cut-point Specificity Sensitivity PPV NPV Max % change by 3 weeks

(n=27) 1.74 0.65 0.80 0.57 0.85

Max % change by 6 weeks (n=36)

21.19 0.87 0.62 0.73 0.80

Max % change by 9 weeks (n=36)

15.49 0.74 1.00 0.68 1.00

Max % change by 12 weeks (n=36)

19.62 0.71 0.92 0.80 0.95

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint

Page 46: A circulating T-cell differentiation marker to predict ... · 13.06.2020  · 38 (K08CA197966) (F. Ito), R01CA188900 (B.H. Segal), R01CA188900 (B.H. Segal), 39 U01CA154967, P50CA159981-01A1

Supplementary Table S3 Prediction performance for study biomarkers, Related to Figure 4E.

Abbreviations: PPV, positive predictive value; NPV, negative predictive value

Baseline CD8+ tumor-infiltrating lymphocytes (low/high vs.

no/minimal infiltration) (n=24)

Baseline tumor mutational burden (≥10.0/Mb vs. <10.0/Mb)

(n=22) PPV 33.3% (5/15) 20.0% (1/5) NPV 55.6% (5/9) 52.9% (9/17)

Sensitivity 55.6% (5/9) 11.1% (1/9) Specificity 33.3% (5/15) 69.2% (9/13) Accuracy 41.7% (10/24) 45.5% (10/22)

.CC-BY-NC-ND 4.0 International licenseavailable under a(which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprintthis version posted June 14, 2020. ; https://doi.org/10.1101/2020.06.13.095844doi: bioRxiv preprint