Melanoma and Vaccines./ Melanoma y vacunas.
 

 

 

Home Page

About Dermagic

Contact Dermagic

Next Edition

Past Editions

Secret X files 

 

Melanoma and Vaccines./ Melanoma y Vacunas. 

************************************
****** DATA-MEDICOS **********
************************************ 
MELANOMA Y VACUNAS
MELANOMA AND VACCINES
**************************************
****** DERMAGIC-EXPRESS No.60 ******* 
****** 09 JUNIO DE 1.999 *********** 
09 JUNE 1.999
**************************************
***************************************

EDITORIAL ESPAŅOL
=================
Hola Amigos de la red. En esta ocasion les traigo una revision sobre el tema MELANOMA Y VACUNAS. Realmente es impresionante la cantidad de estudios que se han hecho para encontrar una vacuna contra el temido melanoma. En estas 36 referencias quedan evidenciados esos intentos por encontrar una vacuna al melanoma. Las referencias datan de los aņos 90 hasta el 99.

Saludos a todos !!!

Bienvenido Dr. Jorge Alvarado (Maracay), a DERMAGIC/EXPRRESS,

Dr. Jose Lapenta R.,,,

EDITORIAL ENGLISH
=================
Hello Friends of the net. In this occasion I bring a revision on the topic MELANOMA AND VACCINES. It is really impressive the quantity of studies that have been made to find a vaccine against the feared melanoma. In these 36 references those intents are evidenced to find a vaccine to the melanoma. The references date of the nineties up to the 99.

Greetings to ALL, !!

Dr. Jose Lapenta R.,,,
=====================================================================
MELANOMA Y VACUNAS / MELANOMA AND VACCINES
=====================================================================
=====================================================================
1.) Active specific immunotherapy with polyvalent melanoma cell vaccine for
patients with in-transit melanoma metastases.
2.) Enhancement of complement-dependent cytotoxicity by polyvalent melanoma
cell vaccine (CancerVax): correlation with survival.
3.) Overview of melanoma vaccines: active specific immunotherapy for
melanoma patients.
4.) Correlation of specific immune responses with survival in melanoma
patients with distant metastases receiving polyvalent melanoma cell vaccine.
5.) TA90 immune complex predicts survival following surgery and adjuvant
vaccine immunotherapy for stage IV melanoma.
6.) Current status of melanoma vaccines.
7.) Vaccines and other adjuvant therapies for melanoma.
8.) Autologous hapten-modified melanoma vaccine as postsurgical adjuvant
treatment after resection of nodal metastases.
9.) Prolongation of survival in metastatic melanoma after active specific
immunotherapy with a new polyvalent melanoma vaccine.
10.) Immunotherapy and experimental approaches for metastatic melanoma.
11.) Allogeneic cells vaccine increases disease-free survival in stage III
melanoma patients. A non randomized phase II study.
12.) Relationship between immune response to melanoma vaccine immunization
and clinical outcome in stage II malignant melanoma.
13.) Increased effectiveness of interferon alfa-2b following active
specific immunotherapy for melanoma.
14.) Active-specific immunotherapy for melanoma.
15.) Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in
association with histocompatibility leukocyte antigen DR11.
16.) A 15-year follow-up of AJCC stage III malignant melanoma patients
treated postsurgically with newcastle disease virus (NDV) oncolysate and
determination of alterations in the CD8 T cell repertoire.
17.) IgM anti-ganglioside antibodies induced by melanoma cell vaccine
correlate with survival of melanoma patients.
18.) Changes in the fine specificity of gp100(209-217)-reactive T cells in
patients following vaccination with a peptide modified at an HLA-A2.1
anchor residue.
19.) Improved efficacy of dendritic cell vaccines and successful
immunization with tumor antigen peptide-pulsed peripheral blood mononuclear
cells by coadministration of recombinant murine interleukin-12.
20.) Development of herpes simplex virus replication-defective multigene
vectors for combination gene therapy applications.
21.) Developing recombinant and synthetic vaccines for the treatment of
melanoma.
22.) New trends in the development of cancer vaccines.
23.) A new mouse model of experimental melanoma for vaccine and lymphokine
therapy. 
24.) Dinitrophenyl-modified autologous melanoma vaccine induces a T cell
response to hapten-modified, melanoma peptides. 
25.) Stimulation of CD8+ T cell responses to MAGE-3 and Melan A/MART-1 by
immunization to a polyvalent melanoma vaccine. 
26.) Double-copy bicistronic retroviral vector platform for gene therapy
and tissue engineering: application to melanoma vaccine development. 
27.) Favorable clinical responses in subsets of patients from a randomized,
multi-institutional melanoma vaccine trial. 
28.) Increased survival of patients treated with a vaccinia melanoma
oncolysate vaccine: second interim analysis of data from a phase III,
multi-institutional trial. 
29.) Active specific immunotherapy with hapten-modified autologous melanoma
cell vaccine. 
30.) IgM anti-ganglioside antibodies induced by melanoma cell vaccine
correlate with survival of melanoma patients. 
31.) Cancer vaccines.
32.) Active specific immunotherapy of metastatic melanoma with an
antiidiotype vaccine: a phase I/II trial of I-Mel-2 plus SAF-m. 
33.) Allogeneic murine melanoma cell vaccine: a model for the development
of human allogeneic cancer vaccine. 
34.) Immune response to polyvalent melanoma cell vaccine in AJCC stage III
melanoma: an immunologic survival model. 
35.) Immunization with a tumor-cell-lysate-loaded
autologous-antigen-presenting-cell-based vaccine in melanoma. 
36.) Augmentation of IgM antibody to gp43 tumor-associated antigen peptide
by melanoma cell vaccine. 
======================================================================
======================================================================
1.) Active specific immunotherapy with polyvalent melanoma cell vaccine for
patients with in-transit melanoma metastases.
======================================================================
Cancer 1999 May 15;85(10):2160-9 

Hsueh EC, Nathanson L, Foshag LJ, Essner R, Nizze JA, Stern SL, Morton DL
Roy E. Coats Research Laboratories, John Wayne Cancer Institute at Saint
John's Health Center, Santa Monica, California 90404, USA. 

BACKGROUND: This study was conducted to document the rate, duration, and
type of objective response to active specific immunotherapy with a
polyvalent melanoma cell vaccine (PMCV) for patients with in-transit
melanoma metastases and to identify any acute or chronic toxic effects of
PMCV treatment. METHODS: An analysis was conducted of all in-transit
melanoma patients seen at the John Wayne Cancer Institute in Santa Monica,
California, during the period 1985-1997 who were enrolled in prospective
PMCV protocols in the absence of other therapies with possible antitumor
activity (n = 54). Clinical response to PMCV was assessed by standard
criteria. Survival curves were estimated by the Kaplan-Meier method.
Toxicity was graded according to the Eastern Cooperative Oncology Group
standard. RESULTS: PMCV produced a 17% (9 of 54 patients) objective
response rate with a 13% rate (7 of 54 patients) of complete remission
(CR). The median duration of CR was >22 months. Complete response lasting
more than 1 year was observed in 4 patients (7.2%); 1 patient remained in
remission over 9 years. Median survival was >53 months (i.e., not reached)
for responders, 42 months for nonresponders, and 53 months overall. Salvage
interventions allowed reinduction with PMCV in 23 of 25 patients, who
subsequently remained clinically free of disease for a median of 14 months.
Overall toxicity was mild, easily tolerable, and did not significantly
change the quality of life. There were no toxic deaths. CONCLUSIONS: PMCV
can cause objective complete regression of measurable intransit metastatic
melanoma with minimal toxicity, and may prolong patients' median survival. 

======================================================================
2.) Enhancement of complement-dependent cytotoxicity by polyvalent melanoma
cell vaccine (CancerVax): correlation with survival.
======================================================================
Ann Surg Oncol 1998 Oct-Nov;5(7):595-602 

Hsueh EC, Famatiga E, Gupta RK, Qi K, Morton DL
Sonya Valley Ghidossi Vaccine Laboratory, John Wayne Cancer Institute at
Saint John's Health Center, Santa Monica, California 90404, USA. 

BACKGROUND: Case control studies have demonstrated that administration of
CancerVax, a polyvalent melanoma cell vaccine (PMCV), after complete
resection of melanoma metastases produces a significant improvement in
disease-free survival (DFS). Because PMCV has no direct cytotoxic effect on
melanoma cells, the authors hypothesized that it prolongs survival by
enhancing antibody-mediated antimelanoma cytotoxicity. METHODS: One hundred
melanoma patients participating in a trial of PMCV adjuvant therapy
following complete resection of regional node metastases were randomly
selected for study. Serum samples obtained immediately before (T0) and 4,
8, 12, and 16 weeks after initiation of PMCV adjuvant therapy were adsorbed
with L-14 lymphoblastoid cells and then tested for in vitro
complement-dependent cytotoxicity (CDC) against M-14 cells, a melanoma cell
line not used in PMCV. CDC was expressed as percentage of total cells (n =
10,000) killed. Survival curves were estimated by the Kaplan-Meier method.
Statistical analysis was performed by the signed rank sum test, Spearman
test, log-rank test, and Cox proportional hazard regression. RESULTS:
Median CDC at T0 was 4.5% (range, 0% to 40%). Within 16 weeks after
initiation of PMCV therapy, CDC had increased in 82 (82%) patients. The
median increase of 7.5% (range, -9% to 39%) represented a highly
significant change (signed rank sum test; P = .0001). At a median follow-up
of 29 months (range, 6 to 92 months), the maximum increase in CDC
(deltaCDC) as a continuous variable was significantly correlated with DFS
(P = .0001). Median survival and 5-year DFS were more than 54 months and
less than 54%, respectively, for patients with deltaCDC > or =10% (n = 44)
but only 7 months and 14%, respectively, for those with deltaCDC <10% (n =
56; P = .0001). Multivariate analysis confirmed deltaCDC as the most
significant independent variable associated with DFS following initiation
of PMCV therapy (P = .0001). CONCLUSION: PMCV therapy greatly enhances
serum CDC against melanoma cells. This enhancement is directly correlated
with DFS following initiation of vaccine therapy. 

======================================================================
3.) Overview of melanoma vaccines: active specific immunotherapy for
melanoma patients.
======================================================================
Semin Surg Oncol 1998 Jun;14(4):328-36 

Ollila DW, Kelley MC, Gammon G, Morton DL
The Roy E. Coats Research Laboratories, John Wayne Cancer Institute at
Saint John's Health Center, Santa Monica, California 90404, USA. 

Although a phase III trial has yet to show a statistically significant
improvement in the disease-free or overall survival of melanoma patients
receiving vaccine therapy, several phase II trials have shown enhanced
disease-free and overall survival of patients who develop a humoral and/or
cellular response to a melanoma vaccine. The challenge of active specific
immunotherapy research is to determine which combination of humoral and
cellular immune responses optimizes clinical outcome and how to monitor the
immune response effectively. This review identifies key components of a
successful melanoma vaccine, discusses new ways to modulate and stimulate
the immune system, and summarizes some of the important clinical trials of
active specific immunotherapy for patients with melanoma. 

======================================================================
4.) Correlation of specific immune responses with survival in melanoma
patients with distant metastases receiving polyvalent melanoma cell vaccine.
======================================================================
J Clin Oncol 1998 Sep;16(9):2913-20 

Hsueh EC, Gupta RK, Qi K, Morton DL
Sonya Valley Ghidossi Vaccine Laboratory of the Roy E. Coats Research
Laboratories of the John Wayne Cancer Institute at Saint John's Health
Center, Santa Monica, CA 90404, USA. 

PURPOSE: The mechanisms that underlie the clinical efficacy of melanoma
vaccines are not well understood. We hypothesized that the type and
strength of the immune response generated by CancerVax (John Wayne Cancer
Institute, Santa Monica, CA), a polyvalent melanoma cell vaccine (PMCV),
might be correlated with its effect on overall survival (OS). PATIENTS AND
METHODS: Seventy-seven patients began PMCV therapy after complete surgical
resection of distant metastatic melanoma. During the first two treatments,
PMCV was administered with bacille Calmette-Guerin (BCG). Blood was drawn
at 0, 2, 4, 8, and 12 weeks to measure serum titers of immunoglobulin G
(IgG) and IgM antibodies against a tumor-associated 90-kd glycoprotein
antigen (TA90) expressed on most melanoma cells, including those of PMCV.
Cellular immune response to PMCV was assessed by delayed-type
hypersensitivity (DTH). General immune competence was assessed by skin
tests to purified protein derivative (PPD), mumps, and candida. RESULTS:
Median follow-up time was 31.5 months. Within the first 12 weeks of PMCV
immunotherapy, there was a significant increase in the anti-TA90 IgM
(P=.0001) and IgG titers (P=.0001), and in the DTH response to PMCV
(P=.0001). Univariate analysis showed that high anti-TA90 IgM titer and
strong PMCV-DTH were associated with improved survival (P=.051 and .0173,
respectively), whereas elevated anti-TA90 IgG was correlated with decreased
survival (P=.0119). Multivariate analysis considering clinical variables
and PMCV immune responses identified anti-TA90 IgM, anti-TA90 IgG, and
PMCV-DTH as significant independent variables influencing survival
following PMCV immunotherapy (P=.0342, .0105, and .0082, respectively).
These responses to PMCV were not correlated with immune responses to BCG
and therefore were not a manifestation of general immune competence for
responses to unrelated antigens. The median survival time and 5-year
survival rate were more than 76 months and 75%, respectively, if both
anti-TA90 IgM and PMCV-DTH responses were strong (> or = 800 and > or = 7
mm, respectively; n=29); 32 months and 36%, respectively, if only one
response was strong (n=35); and 19 months and 8%, respectively, if neither
was strong (n=13) (P < .0001). CONCLUSION: PMCV induces both humoral and
cell-mediated immune responses to melanoma-associated tumor antigens, the
type and strength of which appear to be directly related to its therapeutic
efficacy. 

======================================================================
5.) TA90 immune complex predicts survival following surgery and adjuvant
vaccine immunotherapy for stage IV melanoma.
======================================================================
Cancer J Sci Am 1997 Nov-Dec;3(6):364-70 

Hsueh EC, Gupta RK, Qi K, Yee R, Leopoldo ZC, Morton DL
Roy E. Coats Research Laboratories, John Wayne Cancer Institute, Saint
John's Health Center, Santa Monica, California, USA. 

PURPOSE: Although prognosis remains poor for patients with distant
metastatic melanoma, we have observed significantly prolonged survival in
patients receiving our polyvalent melanoma cell vaccine (PMCV) following
complete metastasectomy for American Joint Committee on Cancer (AJCC) stage
IV melanoma. Clinical prognostic factors specific to this stage IV subgroup
have not been well characterized. We previously reported that the serum
immune complex (IC) level of a 90-kD glycoprotein antigen (TA90) was an
objective predictor of survival and recurrence in patients with early-stage
melanoma. In the present study we correlated the postoperative TA90-IC
level of AJCC stage IV patients prior to adjuvant PMCV therapy with their
duration of subsequent survival. PATIENTS AND METHODS: From October 1,
1984, to December 31, 1995, 125 stage IV patients began PMCV after complete
resection of distant melanoma metastases. One blood sample was obtained
immediately prior to vaccine therapy, and the serum TA90-IC level was
assessed as positive or negative using our double-determinant ELISA.
Disease-free and overall survival were recorded prospectively from the
start of vaccine therapy. The correlation between prevaccine TA90-IC level
and survival was assessed by the log-rank test and Cox proportional hazards
model. RESULTS: Median follow-up after PMCV therapy was 36.5 months, with a
minimum of 12 months. Univariate analysis demonstrated that TA90-IC level
is significant for both overall survival and disease-free survival. Median
overall survival, median disease-free survival, and rate of 5-year survival
were higher for patients with negative TA90-IC levels than for those with
positive TA90-IC level (58 vs 19 months, 7 vs 4 months, and 49% vs 27%,
respectively). Multivariate analysis established TA90-IC as an independent
prognostic indicator for both overall and disease-free survival following
adjuvant PMCV therapy for AJCC stage IV melanoma. CONCLUSION: Prevaccine
TA90-IC level correlated strongly with overall and disease-free survival in
our stage IV melanoma patients receiving postoperative PMCV immunotherapy.
This is the first serum marker shown to have importance in predicting the
survival of melanoma patients receiving adjuvant immunotherapy after
complete resection of distant metastases. 

======================================================================
6.) Current status of melanoma vaccines.
======================================================================
Dermatol Surg 1997 Aug;23(8):649-54; discussion 654-5 

Kuhn CA, Hanke CW
Department of Dermatology, Indiana University School of Medicine,
Indianapolis, USA. 

BACKGROUND: Malignant melanoma is increasing worldwide faster than any
other cancer and the American lifetime risk is estimated to reach 1 in 75
by the year 2000. Active specific immunotherapy with vaccines is evolving
as a promising new modality in the treatment of malignant melanoma.
OBJECTIVE: To present a concise and understandable summary of the key
molecular and clinical concepts of melanoma vaccines currently under
investigation, the history that led to their development, and their
anticipated clinical response. METHODS: The recent advances in the field of
melanoma immunobiology and the newest experiment vaccines are reviewed.
RESULTS: There is no effective melanoma vaccine that successfully treats or
prevents melanoma. However, their use has been associated with regression
or delayed disease progression in some cases. The minority of patients who
do have a major clinical response to vaccine therapy experience an
improvement in survival. Even in those patients in whom melanoma vaccines
cannot improve survival, the paucity of severe side effects has provided a
quality of life superior to standard multiagent chemotherapy. CONCLUSION:
Melanoma vaccines are relatively safe immunotherapeutic modalities for the
management of malignant melanoma. The clinical effectiveness of melanoma
vaccines is unclear and adequately controlled studies need yet to be
performed. Current melanoma vaccines manipulate antigen presentation
networks and combine the best cellular and antibody antitumor immune
response effective in mediating tumor protective immunity; these
combination vaccines hold the most promise. The ideal melanoma vaccine will
ultimately prevent melanoma. 

======================================================================
7.) Vaccines and other adjuvant therapies for melanoma.
======================================================================
Hematol Oncol Clin North Am 1998 Aug;12(4):835-48, vii 

Wolchok JD, Livingston PO, Houghton AN
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York,
New York, USA. 

Patients with thick primary melanomas or regional lymph node involvement
are at high risk of relapse. Investigations of adjuvant therapy over the
past 30 years show only one significantly positive trial employing high
dose interferon-alpha-2b. This is a potentially toxic regimen, therefore,
other better-tolerated forms of adjuvant immunotherapy are being studied.
Recent advances in basic science have led to a better understanding of the
T-cell response to human cancer. This article discusses the background and
current clinical trials of active specific immunotherapies for melanoma,
including peptide and ganglioside vaccines. 

======================================================================
8.) Autologous hapten-modified melanoma vaccine as postsurgical adjuvant
treatment after resection of nodal metastases.
======================================================================
J Clin Oncol 1997 Jun;15(6):2359-70 

Berd D, Maguire HC Jr, Schuchter LM, Hamilton R, Hauck WW, Sato T,
Mastrangelo MJ
Department of Medicine, Thomas Jefferson University, Philadelphia, PA
19107, USA. [email protected] 

PURPOSE: To determine whether treatment with an autologous whole-cell
vaccine modified with the hapten dinitrophenyl (DNP vaccine) is an
effective postsurgical adjuvant treatment for melanoma patients with
clinically evident nodal metastases. PATIENTS AND METHODS: Eligible
patients had regional nodal metastases that were large enough (> or = 3 cm
diameter) to prepare vaccine. Following standard lymphadenectomy, patients
were treated with DNP vaccine on a monthly or weekly schedule. RESULTS: Of
62 patients with metastasis in a single lymph node bed (stage III), 36 are
alive after a median follow-up time of 55 months (range, 29 to 76); the
projected 5-year relapse-free and overall survival rates are 45% and 58%,
respectively. Of 15 patients with metastases in two nodal sites, five are
alive with a median follow-up time of 73 months. An unexpected finding was
the significantly better survival of older patients; the projected 5-year
survival of patients greater than 50 versus < or = 50 years was 71% and
47%, respectively (P = .011, log-rank test). The development of a positive
delayed-type hypersensitivity (DTH) response to unmodified autologous
melanoma cells was associated with significantly longer 5-year survival
(71% v 49%; P = .031). Finally, the median survival time from date of first
recurrence was significantly longer for patients whose subcutaneous
recurrence exhibited an inflammatory response (> 19.4 v 5.9 months; P <
.001). CONCLUSION: Postsurgical adjuvant therapy with autologous
DNP-modified vaccine appears to produce survival rates that are markedly
higher than have been reported with surgery alone. Moreover, this approach
has some intriguing immunobiologic features that might provide insights
into the human tumor-host relationship. 


======================================================================
9.) Prolongation of survival in metastatic melanoma after active specific
immunotherapy with a new polyvalent melanoma vaccine.
======================================================================
Ann Surg 1992 Oct;216(4):463-82 
Published erratum appears in Ann Surg 1993 Mar;217(3):309 

Morton DL, Foshag LJ, Hoon DS, Nizze JA, Famatiga E, Wanek LA, Chang C,
Davtyan DG, Gupta RK, Elashoff R, et al
John Wayne Cancer Institute, Santa Monica, CA 90404. 

A new polyvalent melanoma cell vaccine (MCV) was administered to 136 stage
IIIA and IV (American Joint Committee on Cancer) melanoma patients.
Induction of cell-mediated and humoral immune responses to common
melanoma-associated antigens present on autologous melanoma cells was
observed in patients receiving the new MCV. This was accompanied by
increased activation of tumor-infiltrating lymphocytes. Survival correlated
significantly with delayed cutaneous hypersensitivity (p = 0.0066) and
antibody responses to MCV (p = 0.0117). Of 40 patients with evaluable
disease, nine (23%) had regressions (three complete). From our historical
database of 126 stage IIIA and 1275 stage IV melanoma patients, there were
no significant changes in the natural history of metastatic melanoma during
the past 20 years. Univariate and multivariate analyses demonstrated
prognostic significance for site of metastases (p = 0.0001) and
immunotherapy with the new MCV (p = 0.0001). Overall our new MCV increased
the median and 5-year survival of stage IIIA melanoma patients with
regional soft tissue metastases twofold (p = 0.00024), and stage IV
patients threefold (p = 0.0001) compared with previous immunotherapy and
other treatments. 

======================================================================
10.) Immunotherapy and experimental approaches for metastatic melanoma.
======================================================================
Hematol Oncol Clin North Am 1998 Aug;12(4):877-902, viii 

Atkins MB
Melanoma Program, Beth Israel Deaconess Medical Center, Boston,
Massachusetts, USA. 

This article reviews the clinical investigations involving recombinant
cytokines (either alone or in combination with adoptive immunotherapy,
toxicity reduction agents, or cytotoxic chemotherapy), vaccines, monoclonal
antibodies or antibody conjugates, and gene therapy. These various
approaches are reviewed for their current and potential roles in curing
metastatic melanoma. 

======================================================================
11.) Allogeneic cells vaccine increases disease-free survival in stage III
melanoma patients. A non randomized phase II study.
======================================================================
Medicina (B Aires) 1997;57(4):421-7 

Mordoh J, Kairiyama C, Bover L, Solarolo E
Instituto de Investigaciones Bioquimicas Fundacion Campomar, Buenos Aires,
Argentina. 

The incidence of melanoma is increasing rapidly, and in many cases the
primary tumor is excised after metastatic spreading. In 80% of the cases,
the first metastatic site is in regional lymph nodes (AJCC Stage III).
After excision of these nodes, the patient is clinically disease-free, but
the chances of recurrency vary between 40-80%. Thirty patients with stage
III melanoma were treated in a non-randomized Phase II adjuvant trial with
a vaccine consisting of a mixture of three allogeneic cell lines:
IIB-MEL-J, IIB-MEL-LES and IIB-MEL-IAN (5 x 10(6) cells each). The cells
were irradiated (5,000 cGy) and BCG was used as nonspecific stimulant.
Before each vaccination (72 hr) the patients received cyclophosphamide (300
mg/sqm). The untreated control group was composed of 24 Stage III melanoma
patients. Vaccination started within 60 days after surgery, and patients
received 4 vaccinations, one every 21 days and then 1 every two months
during the 1st year; 1 every three months during the 2nd year, and 1 every
6 months during the 3rd, 4th and 5th years. The treated group was composed
by 19 men (63.3%) and 11 women (36.7%); average age: 47.6 +/- 14.1 years
(range: 16-70 yr). The control group was composed by 18 men (75%) and 6
women (25%); average age 49.8 +/- 14.2 yr (range: 26-73 yr). The median
disease free survival (DFS) calculated according to Kaplan-Meier was 7.0
months in the control group vs 20.0 months in the treated group (p <
0.001). The results of this clinical trial suggest that treatment with
allogeneic cell vaccines increases DFS in stage III melanoma patients. 

======================================================================
12.) Relationship between immune response to melanoma vaccine immunization
and clinical outcome in stage II malignant melanoma.
======================================================================
Cancer 1992 Mar 1;69(5):1157-64 

Bystryn JC, Oratz R, Roses D, Harris M, Henn M, Lew R
Department of Dermatology, New York University School of Medicine, NY 10016. 

The authors investigated whether there was a relationship between the
induction of a delayed-type hypersensitivity (DTH) response to melanoma
vaccine immunization and disease recurrence. They studied prospectively 94
evaluable patients with surgically resected Stage II malignant melanoma who
were immunized to a partially purified, polyvalent, melanoma antigen
vaccine. The DTH response to skin tests to the vaccine was measured before
treatment and at the fourth vaccine immunization. Vaccine treatment induced
a strong DTH response in 29 (31%) patients, an intermediate response in 24
(25%), and no response in 41 (44%). The median disease-free survival (DFS)
of patients with a strong, intermediate, and no DTH response to vaccine
immunization was more than 72 months, 24 months, and 15 months,
respectively. The relationship between an increase in the DTH response and
a prolonged DFS was statistically significant (P = 0.02); clinically
meaningful (the median DFS of patients with a strong DTH response was 4.7
years longer than that of nonresponders); and, by multivariate analysis,
independent of disease severity or overall immune competence. These
findings suggest, but do not prove, that vaccine treatment can slow the
progression of melanoma in some patients. 


======================================================================
13.) Increased effectiveness of interferon alfa-2b following active
specific immunotherapy for melanoma.
======================================================================
J Clin Oncol 1994 Feb;12(2):402-11 

Mitchell MS, Jakowatz J, Harel W, Dean G, Stevenson L, Boswell WD, Groshen S
Department of Medicine, Norris Comprehensive Cancer Center, University of
South California School of Medicine, Los Angeles 90033. 

PURPOSE: To determine whether interferon alfa-2b (IFN-alfa; intron-A,
Schering Corp, Kenilworth, NJ) can induce a remission in patients
previously treated with active specific immunotherapy (therapeutic melanoma
vaccine) without response. PATIENTS AND METHODS: Eighteen patients with
disseminated melanoma who had failed to respond to at least five injections
of Melacine therapeutic melanoma vaccine (Ribi ImmunoChem Research, Inc,
Hamilton, MT) were then treated IFN-alfa after a 4-week interval. IFN-alfa
5 or 6 x 10(6) U/m2 was self-administered three times a week subcutaneously
by melanoma patients for at least 2 months. Computed tomographic (CT) scans
of the chest, abdomen, and pelvis and magnetic resonance imaging of the
brain were performed within 4 weeks before treatment as a baseline, and
then at 2-month intervals during treatment to evaluate response. All 18
patients were HLA-typed before treatment. The frequency of cytolytic T-cell
precursors (pCTL) in the blood had been measured weekly in 13 of the
patients during treatment with Melacine. RESULTS: Eight of 18 patients
(44.4%) had a major objective clinical response induced by IFN-alfa,
including site-specific complete remissions in five. Responses lasted a
median of 11 months. The median survival duration of the responders has not
been reached, and exceeds 32 months. The group as a whole had a median
survival duration of 10.1 months, and nonresponders lived 7.3 months.
Cytolytic T-cell precursors had been increased by immunization in all five
responding patients tested, but also in five of eight nonresponders. There
was no association of response to IFN-alfa with specific HLA phenotypes, in
contrast to our previous results with melanoma theraccine alone.
CONCLUSION: These data suggest an additive effect of active specific
immunotherapy and IFN-alfa on the objective response rate, perhaps through
upregulation of HLA molecules and tumor-associated antigens on the tumor
cell by IFN-alfa, after immunization of the patient by Melacine. This
treatment may have improved survival over that expected in metastatic
melanoma. 

=====================================================================
14.) Active-specific immunotherapy for melanoma.
======================================================================
J Clin Oncol 1990 May;8(5):856-69 

Mitchell MS, Harel W, Kempf RA, Hu E, Kan-Mitchell J, Boswell WD, Dean G,
Stevenson L
Department of Medicine, University of Southern California School of
Medicine, Los Angeles. 

Twenty-five patients with metastatic melanoma were treated with a
therapeutic vaccine ("theraccine") consisting of allogeneic melanoma
lysates and a novel adjuvant, DETOX (Ribi ImmunoChem Research, Inc,
Hamilton, MT). Each patient received 200 antigenic units (20 x 10(6) tumor
cell equivalents) subcutaneously on weeks 1, 2, 3, 4, and 6. Clinical
responses included one complete remission, three partial remissions, and a
long-term (17-month) stability. Two other patients had mixed responses,
with partial remissions of numerous subcutaneous nodules. Sites of
responsive disease included primarily the skin, but ileal, breast, and a
liver metastasis also responded. Removal of residual lesions in patients
with partial remissions, whose other lesions had disappeared during
treatment, led to long disease-free survivals. The median duration of
remission was 17 months, with four of the five responders alive for at
least 24 months after treatment. An increase in precursors of cytolytic T
cells (CTLs) correlated with clinical outcome, when complete, partial, and
mixed responses and long-term stability were considered. The CTLs
recognized melanoma-associated antigens on many cell lines, but not other
types of tumor or normal lymphocytes. Skin-test reactivity to melanoma
antigens and serum antibodies against the melanoma cells was unrelated to
clinical response. Toxicity was minimal, restricted largely to minor
soreness at the site of injection. Only five patients, four of whom were
treated with repeated courses, developed severe granulomas. These results
confirm that active-specific immunization with allogeneic lysates of
melanoma administered with the adjuvant DETOX can induce immunity to
melanoma, and can induce regressions of disease in a proportion of patients
with metastatic disease with little toxicity. 

=====================================================================
15.) Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in
association with histocompatibility leukocyte antigen DR11.
=====================================================================
J Exp Med 1999 Mar 1;189(5):871-6 

Manici S, Sturniolo T, Imro MA, Hammer J, Sinigaglia F, Noppen C, Spagnoli
G, Mazzi B, Bellone M, Dellabona P, Protti MP
Laboratory of Tumor Immunology, Department of Biology and Technology
(DIBIT), Scientific Institute H. San Raffaele, 20132 Milan, Italy. 

In this study we used TEPITOPE, a new epitope prediction software, to
identify sequence segments on the MAGE-3 protein with promiscuous binding
to histocompatibility leukocyte antigen (HLA)-DR molecules. Synthetic
peptides corresponding to the identified sequences were synthesized and
used to propagate CD4(+) T cells from the blood of a healthy donor. CD4(+)
T cells strongly recognized MAGE-3281-295 and, to a lesser extent,
MAGE-3141-155 and MAGE-3146-160. Moreover, CD4(+) T cells proliferated in
the presence of recombinant MAGE-3 after processing and presentation by
autologous antigen presenting cells, demonstrating that the MAGE-3 epitopes
recognized are naturally processed. CD4(+) T cells, mostly of the T helper
1 type, showed specific lytic activity against HLA-DR11/MAGE-3-positive
melanoma cells. Cold target inhibition experiments demonstrated indeed that
the CD4(+) T cells recognized MAGE-3281-295 in association with HLA-DR11 on
melanoma cells. This is the first evidence that a tumor-specific shared
antigen forms CD4(+) T cell epitopes. Furthermore, we validated the use of
algorithms for the prediction of promiscuous CD4(+) T cell epitopes, thus
opening the possibility of wide application to other tumor-associated
antigens. These results have direct implications for cancer immunotherapy
in the design of peptide-based vaccines with tumor-specific CD4(+) T cell
epitopes. 

=====================================================================
16.) A 15-year follow-up of AJCC stage III malignant melanoma patients
treated postsurgically with newcastle disease virus (NDV) oncolysate and
determination of alterations in the CD8 T cell repertoire.
=====================================================================
Mol Med 1998 Dec;4(12):783-94 

Batliwalla FM, Bateman BA, Serrano D, Murray D, Macphail S, Maino VC, Ansel
JC, Gregersen PK, Armstrong CA
North Shore University Hospital-NYU School of Medicine, 350 Community
Drive, Manhasset, New York, USA. 

[Medline record in process]

Background: The development of effective adjuvant therapies for the
treatment of high-risk melanoma patients is critical for the prevention of
metastatic disease and improvement of patient survival. Active specific
immunotherapy has been tested as an adjuvant treatment in numerous clinical
trials with overall limited, but occasionally promising, success rates.
Newcastle disease virus (NDV) oncolysate has been utilized as an adjunctive
immunotherapeutic agent in the postsurgical management of these patients. A
phase II study initiated in 1975 using adjuvant vaccine therapy composed of
allogeneic and autologous human melanoma cells infected with live NDV (NDV
oncolysate) in patients with AJCC stage III melanoma following therapeutic
lymph node dissection has shown >60% survival rate at 10 years with no
adverse effects. Continued long-term analysis of trials with promising
early results as well as assessment of immunologic responses generated in
these patients may result in improved therapeutic decisions for clinical
trials in the future. Materials and Methods: We analyzed the 15-year
survival of patients treated postsurgically with NDV oncolysate in the
phase II study described above. In an attempt to understand the
immunological effects of this treatment, we have also carried out a
comprehensive analysis of the peripheral blood T cell repertoire in these
patients. Results: The overall 15-year survival of this group of patients
is 55%. Previous studies have suggested that improved outcome in patients
undergoing immunotherapy is correlated with increased numbers of
CD8(+)CD57(+) cells. In surviving patients, we observed a striking
oligoclonality in the CD8(+) T cell population in peripheral blood, which
reflects clonal expansions in the CD8(+)CD57(+) subset. Conclusions: The
data suggest that adjuvant vaccination with NDV oncolysates is associated
with prolonged survival of patients with lymph node-positive malignant
melanoma and that CD8(+) T cells may be an important component of
therapeutic efficacy. 

=====================================================================
17.) IgM anti-ganglioside antibodies induced by melanoma cell vaccine
correlate with survival of melanoma patients.
=====================================================================
J Invest Dermatol 1999 Feb;112(2):205-9 

Takahashi T, Johnson TD, Nishinaka Y, Morton DL, Irie RF
Department of Biotechnology Sciences, John Wayne Cancer Institute, Santa
Monica, California 90404, USA. 

Melanoma cells express ganglioside antigens GM3, GD3, GM2, and GD2 on their
surface. This study examined whether immunization with a melanoma cell
vaccine induced anti-ganglioside antibody responses in melanoma patients
and whether these responses were correlated with survival. Sixty-six
patients who had received melanoma cell vaccine immunotherapy after
surgical removal of regional metastatic melanoma were identified.
Cryopreserved serum samples from these patients were used in an
enzyme-linked immunsorbent assay to determine the IgM antibody levels to
GM2, GD2, GM3, and GD3 prior to melanoma cell vaccine treatment and 4 wk
after the first melanoma cell vaccine immunization. All antibody levels
significantly increased by week 4 (p < 0.001 for all four antibodies) and
all increases were significantly associated with survival (anti-GD2, p <
0.001; anti-GM2, p = 0.001; anti-GD3, p < 0.001; anti-GM3, p < 0.001).
Anti-tumor activity of these antibodies was proved using five
representative antibody-positive sera in a complement-dependent
cytotoxicity assay with cultured melanoma cell lines. These studies suggest
that GM2, GD2, GM2, and GD3 expressed by melanoma cells can induce specific
IgM antibodies and that high levels of these antibodies might have a
beneficial impact on survival. 

=====================================================================
18.) Changes in the fine specificity of gp100(209-217)-reactive T cells in
patients following vaccination with a peptide modified at an HLA-A2.1
anchor residue.
=====================================================================
J Immunol 1999 Feb 1;162(3):1749-55 

Clay TM, Custer MC, McKee MD, Parkhurst M, Robbins PF, Kerstann K,
Wunderlich J, Rosenberg SA, Nishimura MI
Surgery Branch, National Cancer Institute, National Institutes of Health,
Bethesda, MD 20892, USA. 

In a recent clinical trial, HLA-A2+ melanoma patients were vaccinated with
a peptide derived from the melanoma Ag gp100, which had been modified at
the second position (g9-209 2M) to enhance MHC binding affinity.
Vaccination led to a significant increase in lymphocyte precursors in 10 of
11 patients but did not result in objective cancer responses. We observed
that some postvaccination PBMC cultures were less reactive with tumor cells
than they were with g9-209 peptide-pulsed T2 cells. In contrast,
g9-209-reactive tumor-infiltrating lymphocyte cultures generally reacted
equally with tumor cells and g9-209 peptide-pulsed T2 cells. To investigate
this difference in T cell reactivity, T cell cloids derived from the PBMC
of three patients vaccinated with g9-209 2M were compared with T cell
cloids isolated from g9-209-reactive TIL cultures. All of the T cell cloids
obtained from TIL reacted with HLA-A2+, gp100+ melanoma cell lines as well
as with g9-209 and g9-209 2M peptide-pulsed targets. In contrast, only 3 of
20 PBMC-derived T cell cloids reacted with melanoma cell lines in addition
to g9-209 and to g9-209 2M peptide-pulsed targets. Twelve of twenty
PBMC-derived cloids reacted with g9-209 and g9-209 2M peptide-pulsed
targets but not with melanoma cell lines. And 5 of 20 PBMC-derived cloids
recognized only the g9-209 2M-modified peptide-pulsed targets. These
results suggest that immunizing patients with the modified peptide affected
the T cell repertoire by expanding an array of T cells with different fine
specificities, only some of which recognized melanoma cells. 

=====================================================================
19.) Improved efficacy of dendritic cell vaccines and successful
immunization with tumor antigen peptide-pulsed peripheral blood mononuclear
cells by coadministration of recombinant murine interleukin-12.
=====================================================================
Int J Cancer 1999 Jan 18;80(2):324-33 

Fallarino F, Uyttenhove C, Boon T, Gajewski TF
Department of Pathology, University of Chicago, IL, USA. 

The well-characterized P815 tumor model was used to optimize anti-tumor
immunization approaches in mice. Tumor peptides derived from antigens P198
or P1A were targeted to antigen-presenting cells (APC) by ex vivo pulsing.
Initial experiments with irradiated pulsed splenic dendritic cells (sDC)
injected weekly in the hind footpads for 3 weeks demonstrated cytolytic T
lymphocyte (CTL) generation in 10-20% of mice. Because of the importance of
interleukin-12 (IL-12) in tumor rejection responses, pulsed sDCs also were
given together with recombinant murine IL-12 (rmIL-12). This strategy
induced peptide-specific CTL in 100% of the mice. The IL-12 had to be
injected in the footpads on days 0, 1 and 2 of each immunization week to
achieve an optimal effect. The improvement seen with the addition of IL-12
prompted examination of other sources of APC. Purified resting B cells,
lipopolysaccharide (LPS) blasts and nonfractionated splenocytes or
peripheral blood mononuclear cells (PBMC) were pulsed with peptide and
administered with the same schedule of rmIL-12. Because these cell types
appeared to bind peptides less avidly than did DC, increasing peptide doses
were used during pulsing. Interestingly, immunization with each of these
APC also induced specific CTL in 100% of mice, provided rmIL-12 was
coadministered. CTLs were detected both in the spleen and in the peripheral
blood. Immunization with irradiated, P1A-pulsed PBMC plus rmIL-12 resulted
in protection against challenge with tumors expressing the specific antigen
in all mice. The ease by which human patient PBMCs can be prepared provides
a straightforward vaccination approach to be used in clinical trials of
peptide-based immunization in melanoma. 

=====================================================================
20.) Development of herpes simplex virus replication-defective multigene
vectors for combination gene therapy applications.
=====================================================================
Gene Ther 1998 Nov;5(11):1517-30 

Krisky DM, Marconi PC, Oligino TJ, Rouse RJ, Fink DJ, Cohen JB, Watkins SC,
Glorioso JC
Department of Molecular Genetics and Biochemistry, University of Pittsburgh
School of Medicine, PA 15261, USA. 

Some gene therapy applications will require simultaneous expression of
multiple gene products to achieve a therapeutic effect. In this study we
describe the generation and characterization of replication incompetent
herpes simplex virus type 1 (HSV-1) vectors (HX86Z or HX86G) carrying
distinct and independently regulated expression cassettes for five
transgenes (hIL-2, hGM-CSF, hB7.1, HSV-tk and lacZ or hIFN gamma). The
transgenes, representing 12 kb of DNA sequence, were recombined into
separate loci of a single mutant virus vector deleted for 11.6 kb of vector
sequences representing portions of nine viral genes, ICP4, ICP22, ICP27,
ICP47, UL24, UL41, UL44, US10 and US11. Deletion of the immediate--early
genes ICP4, ICP22 and ICP27 substantially reduced vector cytotoxicity,
prevented early and late viral gene expression and left intact MHC class I
antigen expression. Simultaneous expression of multiple transgenes was
obtained for up to 7 days in primary human melanoma cells with peak
expression at 2-3 days after infection. The transgenes were chosen for
their potential to function synergistically in tumor destruction and
vaccine gene therapy applications, but the method and vector employed could
be applied to other multigene therapy strategies. This study demonstrates
the potential for engineering large transgene capacity DNA viruses such as
HSV-1 for expression of multiple transgenes. 

=====================================================================
21.) Developing recombinant and synthetic vaccines for the treatment of
melanoma.
=====================================================================
Curr Opin Oncol 1999 Jan;11(1):50-7 

Restifo NP, Rosenberg SA
National Cancer Institute, National Institutes of Health, Bethesda, MD
20892-1502, USA. 

To develop new vaccines for the treatment of patients with cancer, target
antigens presented on tumor cell surfaces have been cloned. Many of these
antigens are non-mutated differentiation antigens and are expressed by
virtually all melanomas, making them attractive components for a widely
efficacious melanoma vaccine. These antigens are also expressed by
melanocytes, however, and are likely to be subject to immune tolerance. A
central challenge for tumor immunologists has thus been the breaking of
tolerance to cancer antigens. We review recent clinical trials using
experimental cancer vaccines, including recent evidence that therapeutic
vaccines can induce objective responses in patients with metastatic
malignant melanoma. We focus on the foundations of these approaches in new
experimental animal models designed to test novel vaccines and report on
what these new models predict for the future development of therapeutic
vaccines for cancer. 

=====================================================================
22.) New trends in the development of cancer vaccines.
=====================================================================
In Vivo 1998 Nov-Dec;12(6):629-38 

Minev BR, Chavez FL, Mitchell MS
Center for Biological Therapy and Melanoma Research, University of
California, San Diego, La Jolla 92093, USA. [email protected] 

Recent advances in understanding of the molecular mechanisms of antigen
processing and presentation, and the identification of tumor-associated
antigens in melanoma and other cancers, have stimulated the development of
a new generation cancer vaccines. This review summarizes the most recent
approaches for the design of safe and more effective vaccines for cancer.
Peptide-based vaccines are safe and can be synthesized with high purity and
reproducibility. Recombinant viruses encoding tumor-associated antigens
allow efficient delivery and precise control over the form and the quantity
of the delivered antigens. DNA-based vaccines induce long-lasting immune
responses and are considered very safe. Antigen-loaded dendritic cells, and
the use of newly developed adjuvants are also very promising new
approaches. In this review, we also discuss the possible clinical
applications and future directions for vaccine development. 


=====================================================================
23.) A new mouse model of experimental melanoma for vaccine and lymphokine
therapy. 
=====================================================================
Author 
Shrayer DP; Bogaars H; Wolf SF; Hearing VJ; Wanebo HJ 
Address 
Surgical Oncology Research Laboratory, Roger Williams Medical Center,
Providence, RI 02908, USA. 
Source 
Int J Oncol, 13(2):361-74 1998 Aug 

Abstract 
The annual incidence of malignant melanoma is estimated at 10-12 per
100,000 inhabitants in countries of central Europe and the United States,
and alarmingly there has been a dramatic upward trend in that estimate. The
B16 murine melanoma is a rapidly growing metastatic tumor of spontaneous
origin, as are human malignant melanomas. Melanoma cells produce specific
antigens which are uniquely different from normal cellular antigens, and
the expression of such antigens is the cornerstone for preparation of
anti-melanoma vaccines. One major problem in evaluating the effectiveness
of vaccination and other biologic therapies is the variability of
experimental tumor models. A new metastatic model of experimental melanoma
which was developed in our laboratory imitates the major clinical stages of
malignant metastatic melanoma: stage I, primary (local) tumor growth and
bone marrow invasion; stage II, regional lymph node involvement; and stage
III, metastasis to distant organs, such as the lungs. This model has been
used successfully for screening vaccines constructed in our laboratory.
Immunization with formalinized vaccines (of extracellular antigens, intact
melanoma cells, or B700 antigen) or irradiated vaccines (of intact melanoma
cells) partially inhibit primary melanoma tumor growth, reduce metastasis
to regional lymph nodes and lungs, and significantly increase mean survival
time. These anti-tumor effects were improved when polyvalent and monovalent
vaccines were combined with IL-2 therapy. We also compared the immunogenic
activity of vaccines made from B16 melanoma cells transfected with genes
encoding murine IL-2 or GM-CSF, and effects on tumor bearing mice were
compared with or without therapy using the corresponding lymphokines. In
sum, comparison of antibody production, growth of primary melanoma tumors,
number of surviving mice, mean survival time, and percent of mice with lung
metastases, showed that the best course of immunotherapy involves
vaccination of mice with irradiated B16 melanoma cells transfected to
secrete GM-CSF, coupled with GM-CSF therapy. 

=====================================================================
24.) Dinitrophenyl-modified autologous melanoma vaccine induces a T cell
response to hapten-modified, melanoma peptides. 
=====================================================================
Author 
Sato T; Bullock TN; Eisenlohr LC; Mastrangelo MJ; Berd D 
Address 
Department of Medicine, Thomas Jefferson University, Philadelphia,
Pennsylvania 19107-5099, USA. 
Source 
Clin Immunol Immunopathol, 85(3):265-72 1997 Dec 
Abstract 
Active specific immunotherapy with dinitrophenyl (DNP)-modified autologous
melanoma vaccine elicits inflammatory responses in metastatic tumor sites.
Postsurgical adjuvant immunotherapy with this vaccine prolongs survival in
stage III melanoma patients. We have reported that, after administration of
DNP-modified melanoma vaccine, T cell responses to DNP-modified autologous
tumor cells are demonstrable in vivo and in vitro. These responses are
hapten specific and MHC restricted. To elucidate this phenomenon, we
investigated the immune response to DNP-modified peptides eluted from
autologous cells. Short peptides were extracted from DNP-modified and
unmodified autologous melanoma cells by an acid elution technique and HPLC
fractionation. Peptides were also extracted from DNP-modified and
unmodified, EB virus-transformed, autologous B lymphoblasts. These various
peptide fractions were loaded onto autologous B lymphoblasts and tested for
ability to elicit a response by a DNP-specific T cell line as measured by
IFN-gamma production. Unexpectedly, stimulatory activity of peptides from
DNP-modified melanoma cells was confined to a single HPLC fraction.
Spectrometric analysis of this fraction confirmed modification of peptides
with DNP. A weaker T cell response was observed to a single HPLC fraction
of DNP-modified peptides from the patient's B lymphoblasts. No T cell
response was elicited by corresponding fractions of peptides eluted from
unmodified melanoma cells or B lymphoblasts. These findings demonstrate the
human T cell response to DNP-modified autologous melanoma cells is mediated
by hapten-modified, MHC-associated peptides. Further investigation of these
peptides could lead to a new strategy for peptide-based cancer 


=====================================================================
25.) Stimulation of CD8+ T cell responses to MAGE-3 and Melan A/MART-1 by
immunization to a polyvalent melanoma vaccine. 
=====================================================================
Author 
Reynolds SR; Oratz R; Shapiro RL; Hao P; Yun Z; Fotino M; Vukmanovi´c S;
Bystryn JC 
Address 
Ronald O. Perelman Department of Dermatology, New York University Medical
Center, New York 10016, USA. 
Source 
Int J Cancer, 72(6):972-6 1997 Sep 17 
Abstract 
A critical requirement for cancer vaccines is that they stimulate CD8+ T
cell responses. In this study, we tested the ability of a polyvalent
melanoma vaccine to induce CD8+ T cell responses to the melanoma associated
antigens MAGE-3 and Melan A/MART-1. Fifteen HLA-A2+ patients with resected
malignant melanoma were immunized with the vaccine s.c. every 2-3 weeks.
CD8+ T cells in peripheral blood reacting to HLA-A2 restricted epitopes on
MAGE-3 (FLWGPRALV) and Melan A/MART-1/(AAGIGILTV) were quantitated using a
filter spot assay at baseline and following 4 immunizations. Vaccine
immunization induced CD8+ T cells reacting to one or both of these peptides
in 9 of the 15 (60%) patients. These cells were CD8+ and HLA-A2 restricted,
as reactivity was abrogated by monoclonal antibodies (MAbs) to CD8 and
class I HLA, but not by anti-CD4. All responding patients remained
recurrence-free for at least 12 months (median 15 months, range 12 to >21
months), whereas melanoma recurred within 3-5 months in non-responders. The
differences in outcome were unrelated to differences in disease severity or
overall immunological competence between responders and non-responders. Our
results demonstrate directly that MAGE-3 and Melan A/MART-1 can stimulate
CD8+ T cell responses in humans, and suggest that these responses are
protective and surrogate markers of vaccine efficacy. 


=====================================================================
26.) Double-copy bicistronic retroviral vector platform for gene therapy
and tissue engineering: application to melanoma vaccine development. 
=====================================================================
Author 
Wiznerowicz M; Fong AZ; Mackiewicz A; Hawley RG 
Address 
Department of Cancer Immunology, Great Poland Cancer Center, Poznan, Poland. 
Source 
Gene Ther, 4(10):1061-8 1997 Oct 
Abstract 
The efficient genetic modification of solid tumors in situ to stimulate
therapeutic immune responses against them is currently under active
investigation, but is not yet possible using existing gene transfer
technologies. Thus, ex vivo/in vivo vaccination strategies have been
proposed in which the patient's tumor is surgically excised, single cell
suspensions are prepared, the therapeutic genes are introduced and then the
gene-modified cells, after being gamma-irradiated, are injected back into
the patient. However, even with high-efficiency gene delivery systems, this
is a labor-intensive process. Moreover, it is often difficult to obtain
sufficient numbers of gene-modified primary tumor cells during short-term
culturing. On the other hand, extended in vitro passaging of primary tumor
explants may alter their immunophenotypic properties. One approach to
overcome these limitations would be to design universal vaccines consisting
of standardized gene-transduced neoplastic cell lines or mixtures of
gene-transduced cell lines to be combined with autologous tumor samples if
available. Melanoma, which is notable for being one of the most immunogenic
human malignancies, represents a cancer where shared tumor-associated
antigens have been identified. We developed and analyzed several different
retroviral vectors for their ability to stably express exogenous genes at
high levels in a panel of melanoma cell lines. All vectors contained a
reporter gene (nlslacZ) encoding beta-galactosidase with a nuclear
localization signal and the neomycin phosphotransferase (neo) gene as
selectable marker. One vector, DCCMV, which carried a bicistronic
nlslacZ-neo transcriptional unit under the control of the human
cytomegalovirus immediate-early promoter in the U3 region of its 3' LTR,
was found to perform consistently better than the other vectors. The DCCMV
vector, which is an extreme example of the double-copy class of retroviral
vectors, was subsequently used to generate melanoma cell lines
constitutively secreting human interleukin-6 or a soluble form of the human
interleukin-6 receptor for potential use in a phase II clinical vaccine
trial for the treatment of melanoma patients. The DCCMV vector design may
also be useful in gene therapy applications where the intent is to implant
polymer-encapsulated cell lines genetically engineered to stably express
high levels of bioactive proteins. 

=====================================================================
27.) Favorable clinical responses in subsets of patients from a randomized,
multi-institutional melanoma vaccine trial. 
=====================================================================
Author 
Wallack MK; Sivanandham M; Whooley B; Ditaranto K; Bartolucci AA 
Address 
St. Vincent's Hospital, Department of Surgery, New York, NY, USA. 
Source 
Ann Surg Oncol, 3(2):110-7 1996 Mar 
Abstract 
BACKGROUND: A phase III, randomized, double-blind, multi-institutional
trial was performed evaluating active specific immunotherapy using vaccinia
melanoma oncolysate (VMO) in the surgical adjuvant setting in patients with
stage II melanoma (UICC staging). The first interim analysis showed no
significant difference in disease-free and overall survival. The data were
further analyzed to identify subsets of patients with improved outcome when
treated with VMO. METHODS: Patients received either VMO or placebo of live
vaccinia vaccine virus (V), once a week for 13 weeks and then once every 2
weeks for an additional 39 weeks or until recurrence. Having stratified
patients according to sex, age, number of positive nodes, tumor thickness,
and clinical stage, data were analyzed for disease-free survival and
overall survival. RESULTS: Male patients showed a 17% difference in overall
survival at 4 years when treated with VMO (p = 0.19). A subset of male
patients < 57 years of age with one to five positive nodes showed a 30%
difference at 4 years with VMO (p = 0.06). Patients with clinical stage I
but pathological stage II disease (both male and female), who had undergone
prophylactic node dissection, showed a 23% difference in survival at 3
years with VMO (p = 0.11). CONCLUSIONS: This subset analysis shows
encouraging survival benefit in certain subsets of patients and an
increasing trend in overall survival. Further follow-up of this phase III
trial from a second interim analysis will be forthcoming. 

=====================================================================
28.) Increased survival of patients treated with a vaccinia melanoma
oncolysate vaccine: second interim analysis of data from a phase III,
multi-institutional trial. 
=====================================================================
Author 
Wallack MK; Sivanandham M; Ditaranto K; Shaw P; Balch CM; Urist MM; Bland
KI; Murray D; Robinson WA; Flaherty L; Richards JM; Rosen L; Bartolucci AA 
Address 
St. Vincent's Hospital and Medical Center/New York Medical College, New
York 10011, USA. 
Source 
Ann Surg, 226(2):198-206 1997 Aug 
Abstract 
OBJECTIVE: The efficacy of vaccinia melanoma oncolysate (VMO) vaccine to
increase overall survival and disease-free survival of patients with
surgically resected International Union Against Cancer (UICC) stage II
melanoma was studied in a phase III, randomized, multi-institutional trial.
SUMMARY BACKGROUND DATA: Phase I and II trials with VMO showed minimal
toxicity and clinical efficacy in patients with melanoma. In a recently
completed phase III VMO trial, the first interim analysis performed in
April 1994 showed an increasing trend in the survival of patients treated
with VMO. The second interim analysis was performed in April 1995. METHODS:
Patients with surgically resected stage II (UICC) melanoma were treated
with VMO (N = 104) or placebo vaccinia vaccine virus (V) (N = 113) once a
week for 13 weeks and then once every 2 weeks for a total of 12 months.
Patients' clinical data were collected as of May 1995 and analyzed for
survival. RESULTS: In this second interim analysis, the mean follow-up time
is 42.28 months. No survival difference was observed between VMO and V
treatments. However, in a retrospective subset analysis, a subset of males
between the ages of 44 and 57 years and having one to five positive nodes
(at 2-, 3-, and 5-year intervals, 13.6%, 15.9%, and 20.3% difference
insurvival in favor of VMO [N = 20] when compared to V [N = 18] [p =
0.037]) and another subset of patients with clinical stage I (at 3- and
5-year intervals, 30% and 7% difference in survival in favor of VMO [N =
20] when compared to V [N = 23], [p = 0.05]) showed significant survival
advantage with VMO. CONCLUSIONS: Although VMO vaccine therapy in surgical
adjuvant setting did not produce a significant survival benefit to all
patients with melanoma, patients from the above two subsets had significant
survival benefit. 

=====================================================================
29.) Active specific immunotherapy with hapten-modified autologous melanoma
cell vaccine. 
=====================================================================
Author 
Sato T 
Address 
Department of Medicine, Jefferson Medical College, Thomas Jefferson
University, Philadelphia, PA 19107-5099, USA. [email protected] 
Source 
Cancer Immunol Immunother, 43(3):174-9 1996 Nov 
Abstract 
We have developed a novel approach to cancer immunotherapy-an autologous
whole-cell vaccine modified with the hapten dinitrophenyl (DNP). This
approach elicits significant inflammatory responses in metastatic sites and
some objective tumor responses. Post-surgical adjuvant immunotherapy with
DNP-modified melanoma vaccine in a setting of micrometastatic disease
produces significant survival prolongation in stage III melanoma patients.
Histologically, the inflammatory responses of the tumor consist of
infiltration by lymphocytes, the majority of which are CD8+, HLA-DR+ T
cells. T cells from these lesions tend to have mRNA for interferon gamma. T
cell receptor analysis suggests that the tumor-infiltrating T cells are
clonally expanded. DNP-modified vaccine also induces T cells in the
peripheral blood, which respond to DNP-modified autologous cells in a
hapten-specific, MHC-restricted manner. Moreover, a T cell line generated
from these lymphocytes responded to only a single HPLC fraction of
MHC-associated, DNP-modified tumor peptides. Since inflammatory responses
in metastases were not consistently associated with dramatic tumor
regression, we considered the possibility of immunosuppression at the tumor
site. We found that mRNA for the anti-inflammatory cytokine, interleukin-10
(IL-10) is expressed in most metastatic melanoma tissues and subsequently
demonstrated that IL-10 protein is produced by melanoma cells. Thus the
efficacy of DNP vaccine could be further enhanced by inhibition of IL-10
production or binding. Finally, we expect these results obtained with
melanoma to be applicable to other human cancers. 

=====================================================================
30.) IgM anti-ganglioside antibodies induced by melanoma cell vaccine
correlate with survival of melanoma patients. 
=====================================================================
Author 
Takahashi T; Johnson TD; Nishinaka Y; Morton DL; Irie RF 
Address 
Department of Biotechnology Sciences, John Wayne Cancer Institute, Santa
Monica, California 90404, USA. 
Source 
J Invest Dermatol, 112(2):205-9 1999 Feb 
Abstract 
Melanoma cells express ganglioside antigens GM3, GD3, GM2, and GD2 on their
surface. This study examined whether immunization with a melanoma cell
vaccine induced anti-ganglioside antibody responses in melanoma patients
and whether these responses were correlated with survival. Sixty-six
patients who had received melanoma cell vaccine immunotherapy after
surgical removal of regional metastatic melanoma were identified.
Cryopreserved serum samples from these patients were used in an
enzyme-linked immunsorbent assay to determine the IgM antibody levels to
GM2, GD2, GM3, and GD3 prior to melanoma cell vaccine treatment and 4 wk
after the first melanoma cell vaccine immunization. All antibody levels
significantly increased by week 4 (p < 0.001 for all four antibodies) and
all increases were significantly associated with survival (anti-GD2, p <
0.001; anti-GM2, p = 0.001; anti-GD3, p < 0.001; anti-GM3, p < 0.001).
Anti-tumor activity of these antibodies was proved using five
representative antibody-positive sera in a complement-dependent
cytotoxicity assay with cultured melanoma cell lines. These studies suggest
that GM2, GD2, GM2, and GD3 expressed by melanoma cells can induce specific
IgM antibodies and that high levels of these antibodies might have a
beneficial impact on survival. 

=====================================================================
31.) Cancer vaccines.
=====================================================================
AU: Durrant-LG
AD: CRC Department of Clinical Oncology, University of Nottingham, City
Hospital, UK.
SO: Anticancer-Drugs. 1997 Sep; 8(8): 727-33
ISSN: 0959-4973
PY: 1997
LA: ENGLISH
CP: ENGLAND
AB: A better understanding of immune recognition of cells has led to
identification of potential new targets on tumor cells. Noticeable
successes in melanoma have been immunization with the GM2 ganglioside
vaccine, and the identification of novel antigens such as MAGE, BAGE and
GAGE recognized by T cells cloned from cancer patients with regressing
disease. However, the unexpected finding that other antigens recognized by
these T cells were overexpressed normal differentiation antigens such as
tyrosinase. Pmel 17 and Melan A have led to vaccines developed against
differentiation antigens expressed in other solid tumors. Monoclonal
antibody, anti-idiotype and antigen based vaccines for colorectal target
antigens 17-1A, CEA and 791Tgp72 are all in clinical development. Similarly
HER2/neu and mucin overexpression in breast cancer represent promising
targets. Mutations in tumor oncogenes or suppressor genes which lead to
malignant transformation can also present tumor-specific antigens. The most
effective vaccines against infectious disease are live viruses. The
development of DNA vaccines which act like viruses in entering cells and
show continuous production of antigens offers great potential for the future.

=====================================================================
32.) Active specific immunotherapy of metastatic melanoma with an
antiidiotype vaccine: a phase I/II trial of I-Mel-2 plus SAF-m. 
=====================================================================
Author 
Quan WD Jr; Dean GE; Spears L; Spears CP; Groshen S; Merritt JA; Mitchell MS 
Address 
Center for Biological Therapy and Melanoma Research, University of
California, San Diego, La Jolla 92093-0061, USA. 
Source 
J Clin Oncol, 15(5):2103-10 1997 May 
Abstract 
PURPOSE: To determine the toxicity and immunologic activity of an
antiidiotype melanoma vaccine that consists of monoclonal antibody I-Mel-2
(MELIMMUNE-2, IDEC Pharmaceuticals, La Jolla, CA) and an immunologic
adjuvant SAF-m. PATIENTS AND METHODS: Twenty-six patients with metastatic
melanoma, 17 of whom had previously received chemotherapy, were given 2 mg
of I-Mel-2 and either 100 micrograms (n = 6) or 250 micrograms (n = 20) of
SAF-m. Antiidiotype vaccine was given intramuscularly (IM) biweekly for 4
weeks, and then bimonthly until disease progression. Human antimurine
antibodies (HAMA), anti-I-Mel-2 antibodies, and specific antibody (Ab)3
against the melanoma epitope mimicked by the vaccine were titrated before
treatment, biweekly from weeks 4 to 12, and every 4 to 8 weeks thereafter.
Computed tomographic (CT) scans of the chest, abdomen, and pelvis and
magnetic resonance imaging (MRI) of the brain were obtained before and
bimonthly during treatment to evaluate responses. RESULTS: Elevated titers
of human antimouse antibodies and anti-I-Mel-2 antibodies were associated
with clinical antitumor effect (P = .02 and P = .05, respectively). Ab3 was
absent in most patients, but was found in the best clinical responder.
Fever, myalgias/arthralgias, fatigue, nausea, and headaches were the most
common toxicities. Grade III myalgias/arthralgias and headaches required
dose reduction of SAF-m in eight patients at the 250-microgram dose. No
treatment-related death occurred. Six patients had an antitumor effect: one
complete response in liver and lung, two minor responses, and three stable
disease. The patient with a complete response has survived nearly 5 years.
CONCLUSION: I-Mel-2 antiidiotype vaccine was safe, tolerated best at the
100-microgram dose of SAF-m, and had immunologic and clinical activity. 

=====================================================================
33.) Allogeneic murine melanoma cell vaccine: a model for the development
of human allogeneic cancer vaccine. 
=====================================================================
Author 
Knight BC; Souberbielle BE; Rizzardi GP; Ball SE; Dalgleish AG 
Address 
Division of Oncology, St George's Hospital Medical School, Tooting, London,
UK. 
Source 
Melanoma Res, 6(4):299-306 1996 Aug 
Abstract 
In an attempt to induce an immune response against tumour antigens, several
groups are transfecting cytokine and other genes into autologous tumour
cells which are given to the patient as a vaccine. This process is
labour-intensive, time-consuming and expensive. Allogeneic cells would
offer a more convenient vehicle for the delivery of cytokines and other
molecules. However, current dogma suggests that MHC-matched cells are a
prerequisite for an effective immune response. Using murine melanoma models
we compared allogeneic and autologous vaccination and showed that the
survival of C56BL/6 mice (H-2b) was prolonged with some degree of
protection achieved against an autologous B15-F10 (H-2b) cell challenge
when the mice were vaccinated with allogeneic K1735-M2 (H-2k) cells but not
when immunized with autologous B16-F10 cells. Both vaccination with live
and irradiated allogeneic cells induced an anti-tumour effect using only
one immunization and no boost or adjuvant. Protection was not observed
after vaccination with another melanoma (S91; H-2d) or with a carcinoma
(A9HT; H-2k). Allogeneic vaccination promoted a cytotoxic cellular response
against both the allogeneic and the syngeneic melanomas. This allogeneic
vaccination model will be useful for studying the underlying mechanisms of
protection, in both pre- and post-challenge settings, as well as for
developing whole cell vaccination systems using genetically modified
allogeneic tumour cells. 

=====================================================================
34.) Immune response to polyvalent melanoma cell vaccine in AJCC stage III
melanoma: an immunologic survival model. 
=====================================================================
Author 
Jones RC; Kelley M; Gupta RK; Nizze JA; Yee R; Leopoldo Z; Qi K; Stern S;
Morton DL 
Address 
Roy E. Coats Research Laboratories, John Wayne Cancer Institute, Saint
John's Hospital and Health Center, Santa Monica, California, USA. 
Source 
Ann Surg Oncol, 3(5):437-45 1996 Sep 
Abstract 
BACKGROUND: Our polyvalent, allogeneic melanoma cell vaccine (MCV) induces
immunoglobulin M (IgM) and immunoglobulin G (IgG) class antibodies to a
90-kDa glycoprotein melanoma-associated antigen (MAA). Additionally, MCV
induces delayed-type hypersensitivity (DTH) responses that we previously
correlated with survival. We hypothesized that early DTH responses to MCV
and early humoral responses to the 90-kDa MAA expressed on MCV cells may be
predictive of overall survival. We tested this hypothesis by monitoring
immunologic profiles in 59 patients with melanoma who were receiving MCV
after surgical resection of regional lymph node or soft-tissue metastases.
METHODS: Blood was drawn before vaccine administration, biweekly for 6
weeks, and then monthly. DTH to MCV was recorded at 0, 2, 4, and 8 weeks of
MCV therapy. Mean antibody titers during the first 6-week interval were
calculated. Changes in DTH were calculated as the difference between peak
and prevaccine values (delta DTH). RESULTS: At a median follow-up of 75.6
months (range 5-138), univariate analysis assigned prognostic significance
to gender (p = 0.046), lymph node involvement (p = 0.024), delta DTH (p =
0.044), mean anti-90-kDa MAA IgG (p = 0.0009), and mean anti-90-kDa MAA IgM
(p = 0.0014). In multifactorial analysis, only the three immunologic
variables significantly impacted survival (p = 0.046, 0.0005, and 0.0053,
respectively). A mathematical model based on delta DTH and mean anti-90-kDa
MAA IgG and IgM titers closely approximated the observed individual and
overall survival rates. CONCLUSIONS: The correlation between overall
survival and initial humoral/cellular immune responses to MCV immunotherapy
may be useful in selecting patients most likely to benefit from prolonged
adjuvant immunotherapy. 

=====================================================================
35.) Immunization with a tumor-cell-lysate-loaded
autologous-antigen-presenting-cell-based vaccine in melanoma. 
=====================================================================
Author 
Chakraborty NG; Sporn JR; Tortora AF; Kurtzman SH; Yamase H; Ergin MT;
Mukherji B 
Address 
Department of Medicine, University of Connecticut School of Medicine,
Farmington 06030, USA. 
Source 
Cancer Immunol Immunother, 47(1):58-64 1998 Sep 
Abstract 
The discoveries of human melanoma-associated antigens in molecular terms
have renewed interest in peptide- or peptide- and antigen-presenting-cell
(APC)-based cancer vaccines. Considering the limited scope of immunization
using defined peptides, we have studied an alternative approach of specific
immunization with tumor-lysate-loaded autologous APC (adherent peripheral
mononuclear cells cultured in 1000 U
granulocyte/macrophage-colony-stimulating factor for 14 days) as a
surrogate vaccine. Seventeen patients (11 with active metastatic disease)
were intradermally immunized with the vaccine in a phased dose escalation
(10(5)-10(7) cells/injection) monthly for 4 months. Thirteen patients
completed all four immunizations showing no toxicity (3 patients had to be
taken off study because of progressive disease and 1 patient went off study
as a result of myocardial infarction due to multi-vessel coronary artery
disease). None has shown any immediate or delayed toxicity attributable to
the immunization and none has shown any evidence of autoimmunity. One
patient showed a partial regression of a subcutaneous nodule. Thirteen
patients are alive after 4+ months to 30+ months (17-month median survival
for the group). Nine patients showed evidence of delayed-type
hypersensitivity at the vaccine sites. Monitoring of biological response in
conventional natural killer or cytolytic T lymphocyte assays with pre- and
post-immune peripheral blood lymphocytes revealed no consistent
differences. The vaccine-infiltrating lymphocytes (VIL) from nine specimens
were adequately expanded following in vitro stimulation with the respective
autologous-lysate-loaded APC for phenotypic and functional analyses. Five
of the nine ex vivo expanded VIL were predominantly CD8+. Evidence of an
antigen-specific CD8+ T cell response (cytotoxicity and/or tumor necrosis
factor production) was detected in three of the five CD8+ VIL. These
observations suggest that this type of vaccine is feasible, that it has
biological activity, and that the approach may be improved through
additional strategic manipulations. 

=====================================================================
36.) Augmentation of IgM antibody to gp43 tumor-associated antigen peptide
by melanoma cell vaccine. 
=====================================================================
Author 
Takahashi T; Conforti A; Kikumoto Y; Hoon DS; Morton DL; Irie RF 
Address 
Department of Biotechnology Sciences, John Wayne Cancer Institute, Santa
Monica, California. 
Source 
J Clin Immunol, 18(4):299-305 1998 Jul 
Abstract 
We previously reported that gp43 tumor-associated antigen peptide
(DLTMKYQIF; designated 810 antigen) on human melanoma cells is recognized
by IgM human monoclonal antibody L92 and by cytotoxic T lymphocytes (CTL).
In this study, we retrospectively tested sera of 44 patients with regional
metastatic melanoma (22 who recurred within 1 year and 22 who survived
longer than 5 years) to determine if antibody responses to 810 antigen
could be induced by immunization with an allogeneic melanoma cell vaccine
that contained 810 peptide. IgM and IgG antibodies were assessed by
enzyme-linked immunosorbent assay using a synthetic 810 nonamer peptide. A
significant augmentation of IgM antibody was demonstrated 4 weeks after
initiation of vaccine therapy, and the IgM level was significantly higher
in patients who survived more than 5 years. The antigen epitope recognized
by antibodies was located within TMKYQI. Of this epitope sequence, K
appears to play a central role in antigenicity. The 810 antigen recognized
by antibody and CTL may have clinical relevance as a potential source of
melanoma vaccine. 
====================================================================
DATA-MEDICOS/DERMAGIC-EXPRESS No (60) 09/06/99 DR. JOSE LAPENTA R. 
====================================================================

 
 
 
Produced by Dr. Jose Lapenta R. Dermatologist 
Maracay Estado Aragua Venezuela 1.999
            Torre Maracay Las delicias 
Telf: 016-6401054- 043327287-043328571