Supplementary Table 1

advertisement
Supplementary Table 1 | Immunotherapies with FDA-approval for cancer treatment
Agent
Class
Indication
FDA
Ipilimumab
Anti-CTLA-4-blocking mAb
Metastatic melanoma
Reference ID: 3417,736
Nivolumab
Anti-PD-1-blocking mAb
Metastatic melanoma, squamous lung cancer
Reference ID: 3677,021
(2nd line)
Pembrolizumab
Anti-PD-1-blocking mAb
Metastatic melanoma
Reference ID: 3621,876
Sipuleucel
Cell-based vaccine
Castration resistant metastatic prostate cancer
SR-1346.02, 28 SEP
(Provenge)
High dose IL-2
2009
Cytokine
Metastatic melanoma, metastatic renal cell
cancer
Abbreviations: CTLA-4, cytotoxic T-lymphocyte protein 4; mAb, monoclonal antibody.
1
Reference ID: 3165,255
Supplementary Table 2 | Clinical trials on cell-based immunotherapies (DCs) in HCC
Reference
Agent
Treatment schedule
Study design
Population
Endpoint
Relevant findings
Chi et al.
Autologous
2 IT injections
Prospective,
14 patients with
Not reported*
RR: 2 PR, 7 SD by WHO criteria;
(2005)S1
immature DCs
2 days after EBRT
single arm,
advanced tumours
increased AFP-specific immune
(8 Gy) separated by
dose finding
suitable for EBRT
responses in 4/10 patients;
21 days
increased NK cell activity in 3/10
patients; no relevant toxicity
Nakamoto
Autologous
1 IA injection
Prospective,
10 patients with HCV-
Not reported*
No difference in RFS compared with
et al.
immature DCs
mixed with
single arm
related tumours in
a historical control group; DC
(2007)S2
pulsed with
Gelfoam, during
TNM stage >II treated
migration to regional lymph nodes
HepG2-derived
selective
with TAE
not observed; ELISPOT response to
tumour lysate
embolization
AFP, hTERT, Her-2/neu and MRP3
increased by 2–20-fold in 6/8
patients; no relevant toxicity
Butterfield
Autologous
3 biweekly ID
Prospective,
16 HLA-A*0201
Safety and
No AFP or tumour responses;
et al.
immature DCs
injections
single arm,
patients with AFP-
immunological
substantial but mostly modest
(2006)S3
pulsed separately
dose finding
expressing TNM stage III
effects*
tetramer responses in 6/10 patients
with 4 AFP
and IV tumours
with concordant ELISPOT responses
peptides
in 5/6; enhanced NK cell
activation; inability to generate
acceptable DCs in 2 patients; no AEs
attributable to vaccination
Tada et al.
Autologous
6 SC injection and
Prospective,
5 patients with early
Safety, feasibility
RR: No tumour response; strong
(2012)S4
mature DCs
topical imiquimod
single arm
and intermediate
and immune
T-cell responses against HCC
pulsed with three
every 2 weeks after
tumours with no prior
activity*
antigens in 5/5 patients
tumour-
TACE
response to TACE
(particularly against AFP and
associated
MAGE-1); no relevant toxicity
proteins AFP,
MAGEA1, GPC3
Mazzolini
Autologous
3 IT injections on
Prospective,
17 patients with
2
P: feasibility and
RR: 0 PR, 1 SD/8 HCC pts; 1/8
et al.
mature DCs
metastatic digestive
safety*
positive DTH with tumour lysate in
(2005)S5
transduced with
tumours (8 patients
S: biological effect
HCC patients; substantial increase
an adenoviral
with advanced HCC)
and antitumour
in tumour infiltration by effector
activity
immune cells in 5 patients with
weeks 0, 3 and 6
single arm
vector encoding
human IL-12
HCC; no relevant toxicity (no DLT)
Qiu et al.
Autologous
2 to 7 IV weekly
Prospective,
9 patients with stage III
(2011)S6
mature DCs
injections (no
single arm
tumours treated by
positive DTH after Tx; prolonged
pulsed with
prespecified
cytoreductive resection
survival compared to a historical
tumour lysate
schedule reported)
followed by RFA,
control group (median 17.1 versus
incubated with
chemotherapy or
10.1 months); increased frequency of
recombinant
interventional Tx
IFN-γ-producing T cells; fever and
Not defined*
bovine α1,3GT,
RR: 3 PR; 0 SD; 8/9 patients had
rash common but not severe
cocultured with
BM-derived CIKs
El Ansary
Autologous
Prospective,
30 patients with
et al.
mature DCs
1 ID injection
randomized
advanced HCC not
reported); increased peripheral CD8+
(2013)S7
pulsed with
(BSC)
amenable to surgery,
T cells; no serious AEs reported
HepG2-derived
Not defined*
RR: 2 PR; 9 SD (criteria not
local ablation or TACE
tumour lysate
Palmer
Autologous
one to six three-
Prospective,
39 patients with
P: tumour response,
RR: 1 PR, 6 SD/25 evaluable
et al.
matured DCs
weekly IV
single arm
advanced HCC not
toxicity
patients; 4 /17 patients had AFP
(2009)S8
pulsed with
injections
amenable to surgery,
S: changes in serum
responses; no relevant toxicity
local ablation or TACE.
AFP and immune
including hepatic toxicity or
response
de novo autoantibody formation
Not reported*
RR: 4 PR, 17 SD/31 patients
HepG2-derived
tumour lysate
Lee et al.
Autologous
Group 1: 5 weekly
Prospective,
31 patients with TNM
(2005)S9
mature DCs
IV injections; group
single arm
stage IV tumours
pulsed with
2: same plus
increased survival in comparison
autologous
monthly injections
with a historical control group; no
tumour lysate
for 2–12 months
relevant toxicity
evaluable; 0/10 DTH responses;
*The method used for sample size calculation was not reported. Abbreviations: α1,3GT, α1,3-galactosyltransferase; AE, adverse events; AFP, α-fetoprotein;
BSC, best supportive care; CIK, cytokine-induced killer cells; DC, dendritic cell; DLT, dose-limiting toxicity; DTH, skin delayed hypersensitivity test; EBRT,
3
external beam radiation therapy; ELISPOT, enzyme-linked immunospot; GPC3, glypican-3; Gy, grey; HCC, hepatocellular carcinoma; Her-2/neu, receptor
tyrosine-protein kinase erbB-2; hTERT, telomerase reverse transcriptase; IA, intra-arterial; ID, intradermal; IT, intrathecal; IV, intravenous; MAGEA1,
melanoma-associated antigen 1; MRP3, canalicular multispecific organic anion transporter 2; NK, natural killer; P, primary endpoint; PR, partial
response; RFA, radiofrequency ablation; RFS, recurrence-free survival; RR, response rate; S, secondary endpoint; SC, subcutaneous; SD, stable disease; TNM,
tumour-node-metastasis; TACE, transarterial chemoembolization; TAE, transarterial embolization; Tx, treatment.
4
Supplementary Table 3 | Clinical trials on cell-based immunotherapies (CIKs) in HCC
Reference
Agent
Treatment schedule
Study design
Population
Endpoint
Relevant findings
Takayama
Autologous
6 IV infusions 2 to
Prospective,
150 patients
P: 3-year RFS and
Increased 3-year RFS (37% versus
et al.
lymphocytes
24 months after
randomized
undergoing R0
TTR
22%) and TTR (median: 2.8 versus
(2000)S10
activated with
resection
resection (76 received
S: disease-specific
1.6 years); no difference in OS; no
CIKs)
survival and OS
relevant toxicity
Clinical effect
No difference in recurrence rates;
rIL-2
Hui et al.
Autologous
3 (group 1) or 6
Prospective,
127 patients with
(2009)S11
lymphocytes
(group 2) IV
randomized
solitary tumours treated
increase in RFS (5-year rate: 23%,
activated with
infusions every
(BSC); sample
by R0 resection (84
19% and 11% in groups 1, 2 and
rIL-2 and rIL-1a
2 weeks after
size
received CIKs)
control); no difference in OS; 5
resection
calculation
patients had DLT (persistent fever)
not reported
Pan et al.
Autologous
At least 4 IV
(2013)S12
lymphocytes
injections every
solitary resected
(5-year OS: 65.9% versus 50.2%);
activated with
2 weeks after
tumours (no selection
multivariate survival analysis
rIL-2 and rIL-1a
resection
criteria for CIK
showed AST, tumour size, tumour
therapy) compared
number, pathological grades and
with a historical
CIK Tx as independent prognostic
Retrospective
204 patients with
OS
cohort
Increased OS in CIK-treated patients
factors
Weng et al.
Autologous
8–10 IV injections
Prospective,
45 patients with
(2008)S13
lymphocytes
given every
randomized
nodular or massive
RFS rate (9% versus 30%); increase
activated with
15 days, starting
(BSC)
HCC (2–13 cm) in
in circulating CD3+, CD4+, and
rIL-2
6–8 weeks after
vascular complete
CD3+CD56+ cells and decreased CD8+
locoregional
response after TACE
T cells; no relevant toxicity
therapy
and RFA Tx
P: TTR and RFS
TTR not reported; reduced 1-year
Ma et al.
Autologous
6 IV infusions
Prospective,
7 patients with small
P: changes in
Increased proportion of CD3+CD8+
(2010)S14
lymphocytes
every 2 weeks
single arm
(1.0–3.5 cm) tumours;
peripheral cell
T cells (36% versus 48.%); increased
activated with
within 3 months
tumour-naive or
subsets and IFN-γ
blood levels of IFN-γ (7.6 versus
rIL-2, and
after RFA
relapsing after prior
levels; tumour
4.7 pg/ml); no recurrence after
therapy
recurrence
7 month follow-up
RetroNectin®
5
(Takara Bio Inc.,
Japan)
Yu et al.
Autologous
2 IV monthly until
Prospective,
132 patients with early
P: OS
Increased OS in CIK-treated patients
(2014)S15
lymphocytes
recurrence (post-
randomized
to advanced tumours
S: PFS
(median 24.9 versus 11.3 months);
activated with
resection),
(BSC)
receiving standard Tx
no difference in recurrence rate or
rIL-2 and rIL-1a
progression (post-
(66 received CIKs)
pattern after resection; no relevant
TACE or BSC) or
toxicity
up to 36 months
Shi et al.
Autologous
3 IV infusions 10
Prospective,
13 patients with mostly
P: Changes in
Increased proportion of CD3+CD8+,
(2004)S16
lymphocytes
to 15 days after
single arm
advanced HBV-related
peripheral
CD3+CD56+ and CD25+ cells and
activated with
apheresis
tumours
lymphocytes subsets
type I and II dendritic cells
rIL-2
Abbreviations: AST, aspartate aminotransferase; BSC, best supportive care; CIK, cytokine-induced killer cells; DLT, dose-limiting toxicity; HCC,
hepatocellular carcinoma; IV, intravenous; OS, overall survival; P, primary endpoint; PFS, progression free survival; R0, negative-margin resection; RFA,
radiofrequency ablation; RFS, recurrence-free survival; S, secondary endpoint; TACE, transarterial chemoembolization; TTR, time to recurrence; Tx,
treatment.
6
Supplementary reference list
S1. Chi, K. H. et al. Combination of conformal radiotherapy and intratumoral injection of adoptive dendritic cell immunotherapy in refractory
hepatoma. J. Immunother. 28, 129–135 (2005).
S2. Nakamoto, Y. et al. Combined therapy of transcatheter hepatic arterial embolization with intratumoral dendritic cell infusion for
hepatocellular carcinoma: clinical safety. Clin. Exp. Immunol. 147, 296–305 (2007).
S3. Butterfield, L. H. et al. A Phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four αfetoprotein peptides. Clin. Cancer Res. 12, 2817–2825 (2006).
S4. Tada, F. et al. Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. Int.
J. Oncol. 41, 1601–1609 (2012).
S5. Mazzolini, G. et al. Intratumoral injection of dendritic cells engineered to secrete interleukin-12 by recombinant adenovirus in patients with
metastatic gastrointestinal carcinomas. J. Clin. Oncol. 23, 999–1010
S6. Qiu, Y. et al. Hepatocellular carcinoma-specific immunotherapy with synthesized α1,3- galactosyl epitope-pulsed dendritic cells and cytokineinduced killer cells. World J. Gastroenterol. 17, 5260–5266 (2011).
S7. El Ansary, M. et al. Immunotherapy by autologous dendritic cell vaccine in patients with advanced HCC. J. Cancer Res. Clin. Oncol. 139, 39–
48 (2013).
S8. Palmer, D. H. et al. A Phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular
carcinoma. Hepatology 49, 124–132 (2009).
S9. Lee, W. C. et al. Vaccination of advanced hepatocellular carcinoma patients with tumor lysate-pulsed dendritic cells: a clinical trial.
J. Immunother. 28, 496–504 (2005).
S10.
Takayama, T. et al. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet
356, 802–807 (2000).
S11.
Hui, D., Qiang, L., Jian, W., Ti, Z. & Da-Lu, K. A randomized, controlled trial of postoperative adjuvant cytokine-induced killer cells
immunotherapy after radical resection of hepatocellular carcinoma. Dig. Liver Dis. 41, 36–41 (2009).
S12.
Pan, K. et al. The efficacy of cytokine-induced killer cell infusion as an adjuvant therapy for postoperative hepatocellular carcinoma
patients. Ann. Surg. Oncol. 20, 4305–4311 (2013).
S13.
Weng, D. S. et al. Minimally invasive treatment combined with cytokine-induced killer cells therapy lower the short-term recurrence rates
of hepatocellular carcinomas. J. Immunother. 31, 63–71 (2008).
S14.
Ma, H. et al. Therapeutic safety and effects of adjuvant autologous RetroNectin activated killer cell immunotherapy for patients with
primary hepatocellular carcinoma after radiofrequency ablation. Cancer. Biol. Ther. 9, 903–907 (2010).
S15.
Yu, X. et al. A randomized Phase II study of autologous cytokine-induced killer cells in treatment of hepatocellular carcinoma. J. Clin.
7
Immunol. 34, 194–203 (2014).
S16.
Shi, M. et al. Autologous cytokine-induced killer cell therapy in clinical trial Phase I is safe in patients with primary hepatocellular
carcinoma. World J. Gastroenterol. 10, 1146–1151 (2004).
8
Download