Personalized Peptide Vaccine for Advanced Pancreatic Cancer



Fig. 40.1
New types of peptide-based vaccines: Examples of new types of peptide-based vaccines are shown. Gray and black boxes indicate CTL and helper T-cell epitopes, respectively





40.2 No Impact of Recent Advances of Cancer Immunotherapy for aPC


Remarkable advances have been made in the field of cancer immunotherapy in the past 5 years. Immune checkpoint blockers (ICB) (e.g., anti-CTLA-4, anti-PD-1, and anti-PDL-1 antibodies) can achieve durable clinical responses in at least one-fifth of patients with several types of advanced cancer [810]. However, these ICB have exhibited very limited clinical benefit for the other cancers, including pancreatic cancer, which displayed no or few tumor-infiltrating lymphocytes [1113]. Cancer vaccines tested in the past two decades also failed to show clinical benefits for pancreatic cancers [1416]. In addition to the dearth of tumor-infiltrating lymphocytes, the large heterogeneity of tumor-associated antigens and the diversity in both HLA and T-cell subsets may hamper the successful development of a cancer vaccine for aPC [35].


40.3 Rationale for Personalized Selections of Vaccine Peptides


Cancer patients possess antitumor immunity, which may depend strongly on both the tumor cell characteristics and the immunological status of the host [1720]. The antitumor immunity differs widely among individuals, since the tumor cell characteristics and the host immune cell repertoires are quite diverse and heterogeneous among patients, even among those with identical HLA types and the same pathological types of cancer. Considering the complexity and diversity of the host immune cell repertoires, it is likely that vaccine antigens that are selected and administered without considering the host immunological status would not efficiently induce beneficial antitumor immune responses [20]. To evaluate the host immune cell repertoires, we examined patients’ pre-existing immunity to a panel of vaccine candidates before vaccination and selected appropriate vaccine antigens for which the individual patients exhibited immunological memory [35]. Vaccine antigens to which patients already possess antigen-specific immunological memory are expected to elicit quick and strong secondary immune responses after vaccination (Fig. 40.2) [35]. In light of this, it would be quite reasonable to select vaccine antigens on the basis of the pre-existing immune cell repertoires in each patient. These facts suggest that our new concept of “personalized” cancer vaccine formulation may confer several advantages, including the possibility of bypassing both immunological diversity and tumor heterogeneity.

A337590_1_En_40_Fig2_HTML.gif


Fig. 40.2
Rationale of personalized peptide vaccine: In conventional peptide vaccines without measuring pre-existing immunity, patients without immunological memory to vaccine antigens would be expected to take more time to develop effective antitumor immune responses, since several rounds of repeated vaccinations might be required to prime antigen-specific naïve T cells to functional effector cells. In personalized peptide vaccines based on pre-existing immunity, patients with antigen-specific immunological memory are expected to show quick and strong secondary immune responses to the selected peptides (This figure is reproduced from reference Sasada et al. [3])


40.4 PPV Procedures


For PPV, a maximum of four peptides are selected based on the results of HLA typing and the pre-existing immune responses specific to each of the 31 HLA class I-restricted cytotoxic T-lymphocyte (CTL) epitope peptides (9-mer or 10-mer short peptides): 12 peptides for HLA-A2, 14 peptides for HLA-A24, 9 peptides for the HLA-A3 supertypes (A3, A11, A31, or A33), and 4 peptides for HLA-A26 (Table 40.1). These peptides were identified mainly through the cDNA expression cloning method with tumor-infiltrating T-lymphocyte lines [35, 2124]. The safety and potential immunological effects of these vaccine candidates have been demonstrated in clinical studies [35, 25, 26]. It should be noted that we currently employ these 31 CTL epitopes, which has also been shown to induce antigen-specific B-cell immune responses, as vaccine antigen candidates for PPV, since it has been suggested that a CTL peptide with the ability to induce antigen-specific B-cell responses could provide more effective immune responses than a CTL peptide without this ability [27, 28].


Table 40.1
Peptide candidates for personalized peptide vaccination










































































































































































































Symbol for peptide

HLA type

Origin protein

Position of peptide

Amino acid sequence

CypB-129

A2,A3sup

Cyclophilin B

129–138

KLKHYGPGWV

Lck-246

A2

p56 lck

246–254

KLVERLGAA

Lck-422

A2,A3sup

p56 lck

422–430

DVWSFGILL

MAP-432

A2,A26

ppMAPkkk

432–440

DLLSHAFFA

WHSC2-103

A2,A3sup,A26

WHSC2

103–111

ASLDSDPWV

HNRPL-501

A2,A26

HNRPL

501–510

NVLHFFNAPL

UBE-43

A2

UBE2V

43–51

RLQEWCSVI

UBE-85

A2

UBE2V

85–93

LIADFLSGL

WHSC2-141

A2

WHSC2

141–149

ILGELREKV

HNRPL-140

A2

HNRPL

140–148

ALVEFEDVL

SART3-302

A2

SART3

302–310

LLQAEAPRL

SART3-309

A2

SART3

309–317

RLAEYQAYI

SART2-93

A24

SART2

93–101

DYSARWNEI

SART3-109

A24,A3sup,A26

SART3

109–118

VYDYNCHVDL

Lck-208

A24

p56 lck

208–216

HYTNASDGL

PAP-213

A24

PAP

213–221

LYCESVHNF

PSA-248

A24

PSA

248–257

HYRKWIKDTI

EGFR-800

A24

EGFR

800–809

DYVREHKDNI

MRP3-503

A24

MRP3

503–511

LYAWEPSFL

MRP3-1293

A24

MRP3

1293–1302

NYSVRYRPGL

SART2-161

A24

SART2

161–169

AYDFLYNYL

Lck-486

A24

p56 lck

486–494

TFDYLRSVL

Lck-488

A24

p56 lck

488–497

DYLRSVLEDF

PSMA-624

A24

PSMA

624–632

TYSVSFDSL

EZH2-735

A24

EZH2

735–743

KYVGIEREM

PTHrP-102

A24

PTHrP

102–111

RYLTQETNKV

SART3-511

A3sup

SART3

511–519

WLEYYNLER

SART3-734

A3sup

SART3

734–742

QIRPIFSNR

Lck-90

A3sup

p56 lck

90–99

ILEQSGEWWK

Lck-449

A3sup

p56 lck

449–458

VIQNLERGYR

PAP-248

A3sup

PAP

248–257

GIHKQKEKSR


A3sup HLA-A3 supertype (A3, A11, A31, and A33), HLA human leukocyte antigen

The safety and immunological effects of these 31 peptides had been confirmed in previous clinical trials, and all peptides were prepared under conditions of Good Manufacturing Practice using a multiple peptide system (San Diego, CA)

For the selection of peptides suitable for each patient, in the earlier stage of translational studies of PPV, pre-existing immunity was defined by the frequencies of CTL precursors in pre-vaccination peripheral blood mononuclear cells (PBMCs) [2933]. However, we are currently evaluating the pre-existing immunity to vaccine candidates by measuring peptide-specific IgG titers in pre-vaccination plasma by the multiplex bead-based Luminex assay rather than CTL precursor frequencies, since the performance characteristics, such as the sensitivity and reproducibility, of the current T-cell assays are sometimes unsatisfactory for detecting low frequencies of antigen-specific CTLs [34, 35].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 26, 2017 | Posted by in ONCOLOGY | Comments Off on Personalized Peptide Vaccine for Advanced Pancreatic Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access