AU682256B2 - Pathogen-specific CTL therapy - Google Patents

Pathogen-specific CTL therapy Download PDF

Info

Publication number
AU682256B2
AU682256B2 AU17725/95A AU1772595A AU682256B2 AU 682256 B2 AU682256 B2 AU 682256B2 AU 17725/95 A AU17725/95 A AU 17725/95A AU 1772595 A AU1772595 A AU 1772595A AU 682256 B2 AU682256 B2 AU 682256B2
Authority
AU
Australia
Prior art keywords
hiv
specific antigen
lymphocytes
human
therapeutic composition
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
AU17725/95A
Other versions
AU1772595A (en
Inventor
Malcolm Gefter
Judy Liebermann
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
New England Medical Center Hospitals Inc
Immulogic Pharmaceutical Corp
Original Assignee
New England Medical Center Hospitals Inc
Immulogic Pharmaceutical Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by New England Medical Center Hospitals Inc, Immulogic Pharmaceutical Corp filed Critical New England Medical Center Hospitals Inc
Publication of AU1772595A publication Critical patent/AU1772595A/en
Application granted granted Critical
Publication of AU682256B2 publication Critical patent/AU682256B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464838Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Virology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Description

Regulation 3.2
AUSTRALIA
Patents Act 1990 COMPLETE SPECW[CAIION FOR A STANDARD PATENT (ORIGIN4AL) 000 0t *000 0 Name of Applicant: New England Medical Center Hospitals, Inc. AND ImmuLogic Pharmaceutical Corporation Address for Service: Invention Title: 1 Little Collins Street, Melbourne, 3000.
"Pathogen-specific CTh therapy' 00 The following statement is a full description of this invention, including the best method of performing it known to us: 1- 950428,q:\oper\1w8M39-91.div,1 1A PATHOGEN-SPECIFIC CTL THERAPY BACKGROUND OF THE INVENTION This invention relates to therapies involving administration of cytotoxic T lymphocytes.
Cytotoxic T cells (CTL's) that specifically lyse HIV-1 infected autologous target cells have been found to occur at uncommonly high frequency in the blood of HIVinfected individuals; killing by such cells is pre ominantly mediated by CD3+CD8 effector cells although cytotoxic CD4 cells and natural killer cells also play a role (Walker et al., Nature 328:345, 1987; Plata et al., Nature 328:348, 1987; Walker et al., Science 240:64, i: 3; Sethi et al., Nature 335:178, 1988; Koenig et al., Proc. Natl. Acad. Sci. USA 85:8638, 1988; Nixon et al., Nature 336:484, 1988; Tsubota et al., J. Exp. Med.
619:1421, 1989; Riviere et al., J. Virol. 63:2270, 1989; Koup et al., Blood 73:1909, 1989; Hoffenbach et al., J.
Immunol. 142:452, 1989; Culmann et al., Eur. J. Immunol.
20 19:2383, 1989; and Hosmalin et al., Proc. Natl. Acad.
Sci. USA 87:2344, 1990). CD8+ T cells recognize antigenic peptides presented by MHC class I molecules.
To be recognized by a CTI a peptide must be properly process.-, be capable of inding to MHC strongly enough 25 to compete with other peptides, and be recognized as a peptide-MHC complex by T cells in the repertoire. Recent studies indicate that in some infections only a small number of peptides meet these criteria and that CTL specific for these epitopes dominate the lytic response 30 (Braciale et al., Immunol. Rev. 98:95, 1987; Whitton et al., J. Virol. 62:687, 1988; Klavinskis et al., J. Virol.
63:4311, 1989; Whitton et al., J. Virol. 62:687, 1988;
I
-2- Braciale et al., Proc. Natl. Acad. Sci. USA 86:277, 1989; Townsend et al., Cell 44:959, 1986).
Hosmalin et al. (1990, supra) and Takahashi et al.
(Proc. Natl. Acad. Sci. USA 85:3105, 1988; Science 246:118, 1989; J. Exp. Med. 170:2023, 1989) report that a small number of epitopes likely dominates the CTL response to HIV-1 in mice. Yamamoto et al. Immunol.
144:3385, 1990) and Miller et al. (Abstract FA 74, Sixth International Conference on AIDS, San Francisco, CA, 1990) report that in SIV-infected macaques, the CTL response to gag appears to have a limited epitope specificity. Culmann et al. (1989, supra) report that, in two human subjects, the CTL response to HIV-encoded nef protein is predominantly directed against the same 16 amino acid region. Koenig at al. Immunol. 145:127, 1990) report that a ten amino acid fragment of the nef protein is recognized by the CTL's of two out of ten HIVseropositive individuals.
SUMMARY OF THE INVENTION :The present application is a divisional application of Australian Application No.
87379/91 the specification of which is herein incorporated by reference.
20 In one aspect, the inventiQn features a method of treating a mammal infected with an intracellular pathogen involving selecting, from a sample of the mammal's lymphocytes, a sub-sample which is enriched for cytotoxic T lymphocytes which recognize a pathogen-specific antigen 25 and which are capable of lysing pathogen-infected cells of the patient, and administering to the patient a therapeutically effective amount of the sub-sample Of cytotoxic T lymphocytes. Preparation of such a pathogenspecific CTL-enriched sub-sample generally involves stimulating the proliferation of a mammal's T-lymphocytes in vitro, using a nonspecific mitogen, in the presence of pathogen-infected cells cells displaying pathogenspecific antigens on their surfaces); inclusion of such pathogen-infected cells results in the selective expansion of a population of CTL's which are capable of
I
3 targeting and lysing host cells harboring the pathogen.
Preparation of the pathogen-specific CTL-enriched subsample may further involve identifying, for a particular mammal, those pathogenic-specific antigens which are capable of eliciting a particularly potent CTL response and presenting peptides displaying thsse antigens to the proliferating T cells in order to further expand the pathogen-specific CTL population in the sub-sample.
In preferred embodiments, the sub-sample is prepared by incubating the sample with a mitogen which is capable of inducing lymphocyte proliferation (preferably, phytohemagglutinin); the sample is further incubated with IL-2; the sample is further ccntacted with a pathogenspecific antigen recognized by CTL's of the mammal and which is capable of inducing a CTL response in the mammal; the method further involves administering to the mammal a therapeutically effective amount of a pathogenspecific antigen recognized by the CTL's of the mammal and which is capable of stimulating a cytotoxic T lymphocyte response in the mammal.
In another aspect, the invention features a method S of treating a mammal infected with an intracellular pathogen involving administering to the mammal a therapeutically effective amount of a pathogen-specific antigen which is recognized by the CTL's of the mammal and which is capable of stimulating a cytotoxic T lymphocyte response in the mammal.
In preferred embodiments of both aspects, the antigen is displayed on the surface of an autologous antigen-presenting cell (preferably a B-lymphocyte); the mammal is a human; the intracellular pathogen is a virus (preferably a human immunodeficiency virus, a human T cell leukemia virus, or a Herpes virus, preferably, an Epstein-Barr virus), a mycobacterium, a protozoan, or a fungus; and the pathogen-specific antigen is
I
_I
4 immunodominant. In the case of a patient infected with a human immunodeficiency virus, the pathogen-specific antigen is preferably an HIV-encoded protein (for example, the product of the env or the pol gene), or a CTL-stimulatory fragment thereof.
By "intracellular pathogen" is meant a diseasecausing organism which resides, during at least a part of its life cycle, within a host cell. By "enriched for cytotoxic T lymphocytes" is meant that the sub-sample has a substantially greater number of pathogen-specific cytotoxic T lymphocytes T lymphocytes which recognize and destroy cells bearing foreign antigens, in this case, pathogen-specific antigens, on their surfaces) than a freshly isolated sample of the patient's lymphocytes. By "pathogen-specific antigen" is meant a protein produced upon infection by a pathogen which is recognized responded to) as foreign by cells, in this case, cytotoxic T cells, of the patient's immune system. By "lyse" is meant to destroy or disintegrate, 20 for example, a host cell harboring a pathogen. By "pathogen-infected cells" is meant those host cells harboring a pathogen, either in an active or a latent state. By "mitogen" is meant a substance that stimulates mitosis and, thus, cell proliferation. By "CTL response" is meant the proliferation of CTL's in response to, and specific for, a stimulatory antigen. By "autologous" is meant occurring in the same patient. By "antigenpresenting cell" is meant any cell capable of displaying on its cell surface an antigen, or an immunogenic fragment thereof. By "human immunodeficiency virus" is meant, without limitation, HIV-1 and HIV-2; By "human T cell leukemia virus" is meant, without limitation, HTLV- I and HTLV-II; by "Herpes virus" is meant, without limitation, Herpes simplex type 1 and type 2, Herpes zoster, and cytomegalovirus as well as Epstein-Barr -il I 5 virus. By "virus" is also meant, without limitation, Papillomavirus, Creutzfeldt-Jakob virus, and feline leukemia virus. By "mycobacterium" is meant, without limitation, Mycobacterium leprae, Mycobacterium tuberculosis. By "protozoan" is meant, without limitation, Toxoplasma gondii, Giardia lamblia, Trypanosoma cruri, organisms of the genus Leishmania, and organisms of the genus Plasmodium which cause malaria.
By fungus is meant, without limitation, Pneumocystis carinii, Candida albicans, and Candida tropicalis. By "CTL-stimulatory fragment" is meant a peptide which is capable of stimulating antigen-specific CTL proliferation. By "immunodominant" is meant (an antigen) capable of eliciting an unusually potent CTL response.
In the method of the present invention, the CTL's of the enriched sub-sample or CTL's expanded in the host following administration of pathogen-specific CTLstimulatory peptide or peptide-bearing antigen-presenting cells recognize and selectively target pathogen-infected 20 cells. Because such pathogen-infected cells represent a small percentage of the total cell population, this method minimizes side effects, such as immunosuppression, which may result from other forms of therapy such as those which destroy or impair the function of host cells which are either infected or at risk of being infected by S the pathogen. Moreover, the pathogen-specific CTL population may be administered to the mammal free of lymphokines, thereby avoiding the vascular-leak syndrome generally associated with lymphokine therapies, at least in humans and mice. Finally, the pathogen-specific CTL population is generated from a mammal's lymphocyte sample. This is an important feature of the invention because antigens capable of inducing an effective CTL response inducing significant CTL proliferation) may vary, and, for example, in the case of HIV-1, do vary ~I-I IIL-l 6 from one individual to the next. By beginning with a mammal's own lymphocytes, it is possible to enrich for CTL's which recognize and lyse cells bearing pathogenspecific antigens which are immunodominant for that particular human or mammal, thereby maximizing a human's or mammal's CTL response to a pathogenic infection.
Other features and advantages of the invention will be apparent from the following description of the preferred embodiments, and from the claims.
DESCRIPTION OF THE PREFERRED EMBODIMENTS The drawing will first briefly be described.
DRAWING
Fig. 1 is a diagrammatic representation of a set of truncated HIV-1 envelope and reverse transcriptase proteins, from which can be derived peptides which are useful as candidate HIV-l-specific, CTL-stimulatory antigens.
Fig. 2 is a schematic representation of the HIV- 1-specific epitopes recognized as immunodominant by the S: 20 CTL's of eight independent HIV-l-seropositive individuals.
EXAMPLE
There now follows a description of a method for generating, from a sample of an HIV-infected patient's 25 lymphocytes, a sub-sample enriched in cytotoxic T lymphocytes (CTL) which recognize an HIV-l-specific antigen and which lyse cells displaying this antigen on their cell surfaces. The method generally involves establishing a T cell line population) from a 30 sample of the patient's lymphocytes and stimulating proliferation of this T cell line with a nonspecific mitogen. Because HIV-1 infected cells cells displaying HIV-1 antigens on their surface) are naturally 14 1~-M
I
7 included in the sample in infected individuals and because the culture conditions enhance viral replication, the final preparation is enriched for cytotoxic T lymphocytes, which recognize and lyse cells displaying an HIV-1 specific antigen. The method may further involve identifying, for a particular patient, an immunodominant HIV-1 specific antigen(s) which is(are) capable of inducing a CTL response and presenting a peptide(s) displaying this epitope to the proliferating T cell line to further, enrich the sub-sample for HIV-1 specific CTL's.
T-Lymphocyte Culture Heparinized whole blood was obtained from HIV-1 infected patients, patients seropositive for HIV-1 as assayed by Western blot analysis, and peripheral blood lymphocytes (PBLs) were isolated by Ficoll-Hypaque density centrifugation. Cells were cultured at 5x10 5 /ml in RPMI 1640 supplemented with 15% fetal calf serum (Hazleton), 2mM HEPES, 2mM glutamine, 100 U/ml 20 penicillin, 100 gg/ml streptomycin, and 50M p- :mercaptoethanol, and T cell lines were established by addition of 2 gg/ml PHA-P (Difco) and 200 U/ml rhu IL2 (Cetus). Twice a week cultures were adjusted to 5x10 5 /ml with fresh IL-2-containing media.
Using this method, T cell lines were generated "from ten CDC group II, five group III and three CDC group o: IV individuals. These cell lines grew vigorously for over a month without further stimulation, even from patients with AIDS, although those cells grew less vigorously. After 6 weeks, most cell lines with no 0further exogenous stimulation had stopped dividing. No T cell line could be generated from one hemophiliac patient with longstanding AIDS and profound immunodeficiency (absolute CD4 count 20/mm 3 CD4/CD8 ratio 0.03).
After 2-3 weeks of culture, T cell lines were screened for cytotoxicity against autologous B cell lines infected with vaccinia virus containing the HIV-1 env gene or a fragment of the pol gene constructs vPE16 and vCF21, respectively, described in Walker et al., 1987, supra; Walker et al., 1988, supra); a B cell line infected with a vaccinia virus containing the lacZ gene was used as a control construct vSC8, described in Chakrabarti et al., Nature 320:535, 1986; Flexner et al., Virol. 166:339, 1988). Autologous EBVtransformed B cell lines were generated from the patient's peripheral blood lymphocyte samples by standard techniques using B95-8 marmoset cell line supernatant.
Env-vaccinia (vPE16)-infected cells expressed the and gpl60 (env) proteins from isolate HIV-lIIIB; polvaccinia (vCF21)-infected cells expressed all but the last 22 amino acids of the reverse transcriptase (RT) protein from isolate HIV-lHxB.
2 Expression of HIV-1 proteins was verified by radioimmunoprecipitation of infected cell lines with aHIV serum as described in Essex et al. (Science 220, 859, 1983). Vaccinia virus was prepared and titered by plaque forming assay on CV-1 cells as described in Mackett, et al. ("The construction and characterization of vaccinia virus recombinants expressing foreign genes", in D. Glover, ed., DNA cloning: A practical approach, Vol II, IRL Press, Oxford, 1985). Env-vaccinia infected cells were, in some cases, also titered by assaying syncytia formation upon cocultivation with C8166 cells (as described in Salahuddin et al., Virology 129, 51, 1983).
For vaccinia virus infection, 1-4 pfu/cell of virus was added to 5x10 5 exponentially growing B cells in 500 ul of media in a 24 well microtiter plate. Cells were incubated, with rocking, at 37 0 C over CO 2 for 30 min; an additional 1 ml of media was added to each well, and 9- 9 cells were further incubated, without rocking, for 16 hr.
Cytotoxicity was measured using a 51 -Chromium (Cr) relc .se assay as follows. B cells target c l s) were pelleted and resuspended in 200ul of serumcontaining media to which was added 100uCi of Na 2 51 CrO 4 (Dupont). After incubation for 1 hr at 37 0 C over CO 2 with occasional mixing, targets were washed 3 times and resuspended at 105 cells/mi. 104 labelled targets were added to triplicate wells of U bottom microtiter plates.
For peptide experiments, the labelled targets were incubated with peptide at a final concentration of gg/ml for 30 min. at 37°C over CO 2 before adding effector cells. Effector cells were suspended at various E:T ratios in 100ul of media and added to target cells; plates were incubated at 37 0 C over CO2 for 4 hr. For each target, spontaneous release (SR) was determined from wells to which 100 ul of media was added, and total release (TR) was calculated from wells containing 100ul 1% NP40. Supernatants (75ul) were collected from each 20 well and were counted on a gamma counter after addition of 75ul of 1% NP40. Percent specific cytotoxicity was calculated from the average cpm as [(average cpm SR)/(TR SR)] x 100. Spontaneous release below 15% of total release was considered acceptable.
S 25 Using such an assay, it was demonstrated that 12 out of 18 PHA-stimulated T cell lines directed significant cytolysis of either RT or env-expressing targets (defined as specific lysis of HIV-1 expressing targets minus lysis of the lacZ expressing target of a, Z:T of 25:1). Five of the ten group II cell lines lysed RT-bearing targets, four lysed env-bearing targets; two of the five group III cell lines lysed RT and env-targets and of the three group IV cell lines, two lysed RT and all lysed env targets. In addition, it was 31 II I 10 demonstrated that such T cell lines directed as much as HIV-1-specific cell lysis above background lacZ target) lysis in a 4 hr assay using an effector:target ratio of 6:1. This was in contrast to autologous, freshly isolated peripheral blood lymphocytes which, from some patients, showed little or no HIV-specific lysis.
Prior to enhanced HIV-l-specific killing, the PHAstimulated T cell lines acquired NK-like activity, presumably in response to the supraphysiological concentration of IL-2; this effect declined over time in culture.
CTL immunodominant peptides To selectively enhance the proliferation of HIV- 1-specific CTL clones, the cultured T cell line was further stimulated with a peptide which included an immunodominant HIV-1-specific epitope recognized by the patient's CTL.
To identify such immunodominant epitopes for a I 20 particular patient, his/her established T cell line was screened after 2-5 weeks in culture for cytotoxicity against autologous B cell targets infected with the vaccinia virus constructs encoding nested truncations of env and RT proteins; cytotoxicity was assayed by 51 Crrelease. The epitope was further defined by assaying cytotoxicity (again, by SCr-release) against autologous targets which presented short overlapping peptides spanning the region identified as immunodominant using truncated proteins. A representative set of vaccinia vectors expressing nested truncations of the above env and RT isolates for env, vPE16, VPE17, vPE18, VPE8, vPE20, vPE21, vPE22; for RT, vCF32-vCF37, described in Hosmalin et al., 1990, supra) are shown in Fig. 1.
Fig. 2 shows that cell lines established from the PBLs of eight independent patients recognize different I s~ls~- Le L9CII1111 ll a~lli~~- 11 immunodominant env and RT epitopes. In particular, in four cell lines studied, response to a single 104 amino acid region amino acids 747 to 851) of the env glycoprotein unequivocally dominated cytolysis; in one cell line, an additional region 1 to 204) was also recognized. To further refine the commonly recognized 104 amino acid epitope, a set of seven 22-amino acid peptides with 8-amino acid overlaps spanning this region was synthesized by standard techniques (by scientists other than the named inventors) and used to sensitize autologous SiCr-labelled targets. Three of the four T cell lines that recognized determinants in this region responded to different peptide epitopes. For one subject, a peptide concentration of 0.2 gg/ml was needed to begin to sensitize targets and the response plateaued at 12 ig/ml. For this subject, three separate T cell lines generated over a period of six months consistently o recognized the same single dominant epitope. The T cell line from another subject, analyzed with a set of amino acid peptides with 10-amino acid overlaps (obtained from the MRC AIDS Reagent Project), also recognized a single peptide amino acids 219 to 238).
Because the HIV-1 isolate used as a source of Scandidate CTL-stimulatory antigens is likely different from the HIV-1 isolates harbored by the patient and because at least some HIV-1 proteins vary considerably in amino acid sequence between isolates, it is likely that viral antigens identified by this method, those described above, are relatively invariant.
Following identification of immunodominant epitopes, peptides bearing these epitopes were used to stimulate CTL proliferation as follows. T cell lines (approximately 3-4 weeks after initiation of culture) were harvested and resuspended in fresh IL-2-containing media at 106 cells/ml. Cell lines were selected with I_ 12 either peptide alone (by adding an equal volume of peptide-containing media to a final concentration of 1- Ag/ml) or peptide presented by autologous antigenpresenting B cells. For the latter, B cell lines were irradiated (5000 rad), resuspended at 106/ml and incubated with peptide at 1-50 pg/ml at 37 0 C with occasional mixing. After 2 hr, the incubated cells were pelleted and added to the T cell line in an equal volume of f"ash media. Twice weekly, T cells were counted and fresh media added to maintain a cellular concentration of 5x10 5 /ml. After 10 days, treated T cell lines were tested for cytotoxicity against vaccinia-infected targets.
In one specific example, T cell line 132, derived from a patient with generalized lymphadenopathy, was shown to recognize both an N-terminal and a C-terminal o epitope in env but none in RT. To selectively enhance theO growth of HIV-1-specific CTL clones with peptide, a three week T cell line from this individual was incubated S 20 either directly with the C-terminal env peptide that it recognized amino acids 802-823 of sequence, YWWNLLQYWSQELKNSAVNLLN) or with irradiated autologous B cells preincubated with this peptide and washed to remove unbound peptide. In both instances, there was substantial enhancement of HIV-1 specific cytotoxicity to the extent that the killing curves for the peptideselected cell lines resembled those obtainable from envspecific clones. Peptide concentrations of 1 gg/ml (350 nM) and 50 ug/ml (1.75 uM) were equally effective; the lower concentration fell on the linear part of the peptide dose response curve for sensitizing targets for cytotoxicity; the higher concentration fell on the plateau. The threshold for a response was 0.2 gg/ml or Cell lines stimulated with peptide grew less well than untreated cells, possibly because of lysis of I iil- C- 13 peptide-presenting T cells; the decline in cell numbers was greater at the higher peptide concentration. Cell lines treated with peptide-presenting cells experienced a dramatic burst of cell growth and increased in number 8fold above untreated cells. They also developed cytotoxicity against EBV-transformed autologous B cells.
Therapy Sub-samples enriched for pathogen-specific CTL's are administered to a pathogen-infected patient as follows. Cells are washed with PBS to remove culture medium and are infused back into the patient by the standard techniques developed for cancer therapy by Rosenberg (see, Rosenberg et al., N. Eng. J. Med.
319:1676, 1988). Typically, infusion is performed S 15 intravenously using 10 9 10 11 cells, and the procedure takns approximately 30 minutes. If necessary, treatment can e repeated. Therapy can be administered soon after pathogen infection or upon onset of symptoms. In addition, one or more PBL samples isolated from a 20 pathogen-infected, asymptcmatic individual, or a CTLenriched sub-sample prepared following pathogen nfection, may be stored, frozen in liquid nitrogen, antil such time as that patient requires therapy, Because the CTL's of the en: zched sub-sample 25 recognize and selectively target pathogen-infected cells and because such pathogen-infected cells represent a small percentage of the total cell population, this method minimizes side effects resulting from generalized cell damage. In the specific example of an HIV-infected pat nt, the enriched CTL sub-sample would target HIVinfected CD4 lymphocytes, monocytes and macrophages, leaving other cells of the immune system (including uninfected CD4-bearing lymphocytic and monocytic cells) intact and thus reducing the risk of immunosuppression.
av-. L 14 This method also avoids the side effects, the vascular-leak syndrome associated with lymphokine therapy.
CTL-stimulatory pathogen-specific antigens peptides including immunodominant epitopes) are administered in therapeutic amounts to a pathogeninfected patient either as a purified peptide or as a processed antigen presented on the surface of an irradiated autologous antigen-presenting cell irradiated.autologous B cells or autologous PBLs, incubated with peptide antigen as described in the above example). Because such CTL-stimulatory pathogenspecific antigens expand CTL populations which recognize and selectively lyse pathogen-infected cells, this method minimizes generalized cell damage. Typically, such a peptide antigen would be mixed with a pharmaceutically acceptable carrier physiological saline) and administered to a patient by the standard procedures, intravenous injection. Alternatively, irradiated antigen-presenting cells would be infused back into a patient by the standard techniques of Rosenberg (supra) as described above. Such CTL-stimulatory pathogenspecific antigens may be administered (as described above) at any time following infusion with a pathogenspecific CTL-enriched sub-sample to further stimulate the pathogen-specific CTL response.
When appropriate, lymphokines such as IL-2 or IL- 4 may be co-administered with either pathogen-specific CTL-stimulatory peptides or sub-samples of pathogenspecific CTL-enriched lymphocytes to further enhance lymphocyte proliferation. To minimize the side effects often associated with this treatment, a patient may be treated with antihistamines, aspirin or acetaminophen, prior to administration of lymphokines. In addition, a patient may be treated with cyclophosphamide prior to i I- st p ~sl 1~11~1~9~A C- I 15 administration of pathogen-specific CTL-stimulatory peptides or pathogen-specific CTL's.
Pathogen-specific CTL's can combat pathogen infection by recognizing and lysing cells infected with pathogen, thereby preventing further spread of infection.
Moreover, certain pathogen-specific CTL's, CTL's specific for Epstein-Barr virus, can be used to prevent or to treat a virus-induced lymphoma in a patient infected with EBV alone or in a patient infected with EBV and a human immunodeficiency virus.
OTHER EMBODIMENTS Other embodiments are within the following claims.
For example, PBLs may be grown in human serum-containing medium or, alternatively, in serum-free medium AIM V, Gibco). Mitogens other than PHA concanavalin A, anti-CD3 monoclonal antibody, or anti-T cell receptor monoclonal antibody) and lymphokines other than IL-2 IL-4) may be used to stimulate lymphocyte proliferation. Any expression vector capable of transfecting or infecting an antigen-presenting cell 'iay S. be used in this invention.
CTL-stimulatory antigens may be included in HIV- 1-encoded proteins other than env and RT, they may be included in the gag or nef proteins. To identify such antigens in gag, nef, or in any HIV-l-encoded protein, a complete set of candidate peptides would be prepared by fragmenting HIV-1 cDNA, cloning each fragment into a vaccinia expression vector (or any appropriate expression vector as defined above), and testing for CTL-stimulatory capability as described in the above example.
Alternatively, candidate peptides may be synthesized in vitro and tested for CTL-stimulatory activity as described in the above example. An epitope domain could be further refined by expressing sub-fragments of the cloned DNA or by synthesizing sub-fragments of the p--l R1-B 1 I 16 candidate peptides, which span the immunogenic region.
Large numbers of fragments could be tested simultaneously by attaching candidate peptides to a series of microtiter wells, adding 51 Cr-labelled autologous target cells, adding an aliquot of a patient's T cells to each well, and screening for cytotoxicity. Fragments shown to encode a CTL-stimulatory peptide would be administered directly to the patient (as a peptide or presented on the surface of an irradiated, autologous antigen-presenting cell) or would be used to selectively expand an HIV-1specific CTL population in a sample of a patient's peripheral blood lymphocytes. Any isolate of HIV-1 may be used as a source of candidate viral-specific antigens, and a patient infected with any isolate of HIV-1 15 HIV-1M) may be treated using the methods of this invention. Proteins or protein fragments homologous to HIV-l-encoded proteins may also be useful in this invention if such proteins or fragments elicit an HIV-1specific CTL response; such proteins may be coded for by other primate lentiviruses, HTLV-I and HTLV-II as well as the simian immunodeficiency viruses. T cell lines or pathogen-infected patients may be presented simultaneously with more than one pathogen-specific CTLstimulatory epitope; such epitopes may be resident in the same or in different proteins.
Similarly, any pathogen-infected mammal (particularly, domesticated animals and livestock) may be treated with a therapeutic amount of a CTL-stimulatory pathogen-specific antigen or with a sub-sample of the mammal's lymphocytes enriched for CTL's which recognize and lyse cells bearing such an antigen, using the methods described above. Moreover, these methods can be used to identify CTL-stimulatory antigens of, and to treat human patients or mammals infected with, other pathogenic viruses including, but not limited to, HIV-2, human T- I I I s -pl -1 CI ~LI ll] tllls~Nb~suc~sssssssssssss~ 17 cell leukemia viruses, Herpes viruses Epstein- Barr virus) as well as any intracellular disease-causing mycobacterium, protozoan, or fungus. Generally, subsamples of pathogen-specific CTL's would be prepared as described above for HIV-, by culturing a patient's lymphocytes in the presence of pathogen-infected cells or in the presence of one or more CTL-stimulatory pathogenspecific epitopes. In the case of an intracellular pathogen which does not reside in peripheral blood lymphocytes, a sample of the patient's or mammal's cells which harbor pathogen would be isolated by standard techniques and co-cultured with a sample of the patient's or mammal's lymphocytes to produce a pathogen-specific CTL-enriched lymphocyte sub-sample.
Pathogen-specific antigens may be presented to T cells in processed form on the surface of an antigenpresenting cell other than a B-lymphocyte, on the surface of an autologous PBL or an autologous cell of monocytic lineage.
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers.
I b -~saa Ai"

Claims (1)

18- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: 1. A therapeutic composition for treating a given human infected with human immunodeficiency virus wherein the composition is produced by: identifying an immunodominant HIV-specific antigen for said human and contacting an isolated subset of said human's lymphocytes with said immunodominant HIV-specific antigen to enrich said subset for cytotoxic T lymphocytes that recognize an HIV- specific antigen and which lyse HIV-infected cells of said human, said infected cells bearing said immunodominant HIV-specific antigen that is recognized by said cytotoxic T lymphocytes. 2. A method for preparing a therapeutic composition for a given human comprising: identifying an immunodominant HIV-specific antigen for said human and contacting an isolated subset of said human's lymphocytes with said iwtonodominant HIV-specific antigen to enrich said subset for cytotoxic T lymphocytes thb i~,'dvely target and lyse HIV-infected cells of said human, said infected cells bearing said immunodominant HIV-specific antigen that is recognized by said cytotoxic T lymphocytes. 3. The combination of the therapeutic composition of claim 1 and an HIV-specific antigen for use in the treatment of a mammal infected with HIV, said cytotoxic T :lymphocytes recognizing said HIV-specific antigen and lysing cells of said mammal infected with HIV. 4. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 747 to 851 of the HIV envelope protein, or an epitope thereof. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 1 to 204 of the HIV envelope protein, or an epitope thereof. I P:\OPERULR\17725.95.CLM 14/7/97 -19- 6. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 802 to 823 of the HIV envelope protein, or an epitope thereof. 7. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 844 to 863 of the HIV envelope protein, or an epitope thereof. 8. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 788 to 809 of the HIV envelope protein, or an epitope thereof. 9. The therapeutic composition of claim 3, wherein the HIV-specific antigen has a sequence corresponding to amino acids 219 to 233 of the HIV envelope protein, or an epitope thereof. The therapeutic composition according to claim 1 or 4 or 5 or 6, or 7 or 8 or 9, or the combination according to claim 3, or the method according to claim 2, substantially as hereinbefore described with reference to the Figures and/or Examples. DATED this 30th day of June 1997 oV New England Medical Center Hospitals, Inc. AND ImmuLogic Pharmaceutical Corporation By DAVIES COLLISON CAVE S" Patent Attorneys for the applicant *o0* i ABSTRACT The present invention provides various compositions, antigens and methods concerning treatment of intracellular pathogens, particularly HIV. fe 0 696:9 o 0@ 0:0 see* a s 950428,p:\oper\miw,87379-div.clin,22
AU17725/95A 1990-09-06 1995-04-28 Pathogen-specific CTL therapy Expired - Fee Related AU682256B2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US57882890A 1990-09-06 1990-09-06
US578828 1990-09-06

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU87379/91A Division AU660422B2 (en) 1990-09-06 1991-09-06 Pathogen-specific CTL therapy

Publications (2)

Publication Number Publication Date
AU1772595A AU1772595A (en) 1995-07-06
AU682256B2 true AU682256B2 (en) 1997-09-25

Family

ID=24314491

Family Applications (1)

Application Number Title Priority Date Filing Date
AU17725/95A Expired - Fee Related AU682256B2 (en) 1990-09-06 1995-04-28 Pathogen-specific CTL therapy

Country Status (2)

Country Link
AU (1) AU682256B2 (en)
FI (1) FI930963A0 (en)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
TAKAHASHI. H. ET AL (1988) ANAS. 85 3105-3109 *
TOWNSEND. A.R.M. ET AL (1986) CELL, 44 959-968 *
WATARI, E. ET AL (1987) JOURNAL OF EXPERIMENTAL MEDICINE *

Also Published As

Publication number Publication date
AU1772595A (en) 1995-07-06
FI930963A (en) 1993-03-04
FI930963A0 (en) 1993-03-04

Similar Documents

Publication Publication Date Title
Yasutomi et al. Immunization with recombinant BCG-SIV elicits SIV-specific cytotoxic T lymphocytes in rhesus monkeys.
Kanai et al. IL-15 stimulates the expansion of AIDS virus-specific CTL.
Moris et al. Dendritic cells and HIV-specific CD4+ T cells: HIV antigen presentation, T-cell activation, and viral transfer
Kornfeld et al. Lymphocyte activation by HIV-1 envelope glycoprotein
Castro et al. Suppression of human immunodeficiency virus replication by CD8+ cells from infected and uninfected chimpanzees
Cameron et al. Dendritic cells exposed to human immunodeficiency virus type-1 transmit a vigorous cytopathic infection to CD4+ T cells
KEEFER et al. Safety and immunogenicity of Env 2-3, a human immunodeficiency virus type 1 candidate vaccine, in combination with a novel adjuvant, MTP-PE/MF59
Shen et al. Recombinant virus vaccine-induced SIV-specific CD8+ cytotoxic T lymphocytes
Jacobson et al. HTLV‐I‐specific cytotoxic T lymphocytes in the cerebrospinal fluid of patients with HTLV‐I‐associated neurological disease
Lusso et al. Cell-mediated immune response toward viral envelope and core antigens in gibbon apes (Hylobates lar) chronically infected with human immunodeficiency virus-1.
DE69420034T2 (en) SYNTHETIC MIXING PEPTIDES THAT GENERATE NEUTRALIZING ANTIBODIES AND THE EFFECT OF CYTOTOXIC T-LYMPHOCYTES AGAINST HIV
Chehimi et al. In vitro infection of natural killer cells with different human immunodeficiency virus type 1 isolates
Yasutomi et al. Synthetic peptide in mineral oil adjuvant elicits simian immunodeficiency virus-specific CD8+ cytotoxic T lymphocytes in rhesus monkeys.
Littaua et al. A CD4+ cytotoxic T-lymphocyte clone to a conserved epitope on human immunodeficiency virus type 1 p24: cytotoxic activity and secretion of interleukin-2 and interleukin-6
AU660422B2 (en) Pathogen-specific CTL therapy
De Maria et al. CD8+ cytolytic T lymphocytes become infected in vitro in the process of killing HIV‐1‐infected target cells
CA2265885A1 (en) Pharmaceutical compositions for the treatment of immune disorders
AU769538B2 (en) Efficient ex vivo expansion of CD4+ and CD8+ T-cells from HIV infected subjects
Zagury et al. One-year follow-up of vaccine therapy in HIV-infected immune-deficient individuals: a new strategy
Lee et al. Cytotoxic activity against maedi-visna virus-infected macrophages
AU682256B2 (en) Pathogen-specific CTL therapy
WO1993014789A1 (en) Pathogen-specific ctl therapy
WO1993003762A1 (en) Immunostimulation
EP0260714B1 (en) The use of activated t-lymphocytes for preparing a pharmaceutical composition for the treatment of AIDS
US6197583B1 (en) Therapeutic compounds