CA1323303C - Advanced anticancer therapy and cytotoxic medicaments for its implementation - Google Patents

Advanced anticancer therapy and cytotoxic medicaments for its implementation

Info

Publication number
CA1323303C
CA1323303C CA000543912A CA543912A CA1323303C CA 1323303 C CA1323303 C CA 1323303C CA 000543912 A CA000543912 A CA 000543912A CA 543912 A CA543912 A CA 543912A CA 1323303 C CA1323303 C CA 1323303C
Authority
CA
Canada
Prior art keywords
egf
cytotoxic
cells
formula
compound
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
CA000543912A
Other languages
French (fr)
Inventor
Daniel Bichon
Andre E. Myers
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.)
Intracel Corp
Battelle Memorial Institute Inc
Original Assignee
Intracel Corp
Battelle Memorial Institute Inc
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 Intracel Corp, Battelle Memorial Institute Inc filed Critical Intracel Corp
Application granted granted Critical
Publication of CA1323303C publication Critical patent/CA1323303C/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
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/642Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

ABSTRACT
Cytotoxic targeted drug polymers comprising a backbone of polyglutamic or polyaspartic acid or copolymers threreof having part of the .gamma. -carboxy-late groups bound to daunomycin or other antitumor agents and conjugated with epidermal growth factor and/or other antibodies and internalizing factors as a targetting device.

Description

1 3 2 ~ ~, IJ v ADv~NrEn ANTICANOER THERAPY AND CYTOTOXIC MEDICAMENTS
FOR ITS IMPLEMENTATION

r~
R~U~ N

The present invention deals with anti-cancer therap~ and, more parti-cularly, concerns a new metnod of combat.ing ~alignar.t di_eases by adminis-tering a medicamen~ ~hich ~will preferentially ~ill cancer cell and have oniy a limited, or insignificant detrimental effect on ;~ealthy cells.
The invention also concerns new selective cytotoxic compounds for impiementing the method, i.e. conjugates consisting of three main covalen-tly bound components: a seiective tumor targetting vector, a biodegradable poly~er backbone acting as an endocellular drug release carrier and leavir.g no toxic residue after resorption and a cytotoxic drug labei especially designed to inhibit cellular growth or to kill malignant cells.
The invention also concerns a method for manufacturlng the nelA medica-ment compounds.

T~E PRIOR ~T

It is ~ell ~nown ~hat current cancer therapy involves the use or ant;mitotic drugs such as adriamycin, vinchristine, bombesin, daunomycin and metothrexate which all have strong undesirable side-effects on the normal cells of the patient. It is therefore important that the activity of antitumor drugs be specifically directed to the malignant cells and have little toxic effect on the normal cells.
Many targeted cytotoxic agents have already been reported based on anti~odies recognizing speciric cell surface antigens coupled to radio-active isotopes with cytotoxic properties, or to toxins such as auromycin, hematoporphyrin, abrin, ricin, diphteria toxin, pseudomonas, exotoxin, gelonin or to the above-mentioned antimitotic drugs. For reviews see P.E.
THORPE et al., (1985), Monoclonal Antibodies 84; Biological & Clinical Applications; Ed. A. RIMCHER~ e; al., Complexes o~ antibodies and antiviral agents, such as interferon have also been tried ~New Scientist ref. April 17, lg86). Further US-A-4,455,985 discloses the binding of Pseudomonas exotoxin to antibodies, for instance antibodies to s?ecific human cell receptors such as the transferrin receptor.

1 3233~
Slowing the release of anticancer drugs by firs, link ng to d sui'able polymer carrier which will then graduall/ rree th* drug and kee? its con-centration effect ve for a longer time at ~.he ~ar~et site const-~ates an improved approach to the problem. For instance W.A.R. VAN HEESE'~lJK et al., (19~5) J. of Control Release 1, 310-15 discloses the binding of anthracy-clin to a polymer which slowly releases the drug and thus maintains a constant concentration thereof in the blood of treated patlents. In this connection, endocellular drug-releasing systems are particularly interes-ting, for instance antimitotic drugs coupled to the side-,-hain of poly-glutamic acid. Indeed, these side-chains are slowly degraded in the presen-ce of y -glut~myl-transferase, an endocellular enzyme ~hich is particularly abundent in tumor cells as compared to normal cells and, consequently, releasing the anti-mitotic drug within the targeted cells. Thus Y. KATO et al., (1984) in J. Medicinal Che. 27. 1602-7, reports bir.ding Daunomycin (~M) to polyglutamic acid (PLGA) and coupling the resulting c~/totoxl-polymer with rat ~ -fetoprotein ( ~-~FP) antibody. Upon trial, the cytotoxic activity of the resulting ~ -AFP-PLGA-DM conjugate was shown to be more effective than nIg (a control antibody), ~ -A~P, unconjugated DM, PLGA-DM or nIg-PLGA-DM.
EP-~ ,720 (TEIJIN) discloses a conjugate comprising an i.~moglobulin capable of binding selectively to a particular antigen possessed by a cell to be killed, a polymer carrier and a cytotoxic substance lin~ed thereto, for instance p-(N,N-bis(2-chloroethyl))-phenylenediamine, Melphalan, 1-( ~-D-arabino-furanosyl)-cytosine and its phosphate, methotrexate, actinomycin D, mitomycin C and the li~e.
US-A-4,485,093 discloses an immunotoxin conjugate for treating mali-gnant deseases, which consists of arsanilic acid and tumor specific anti-odies covalently bound to a poiyslutamic acid bac~-bone.
In the aforesaid approach~ selection of an effective antigen or rece-ptor to target the drug is of prime importance. However, even more impor-tant is the possibility of causing the medicament to penetrate into tne target cell, i.e. to be internalized therein at suitables sites so that, by fast degradation of the back-bone drug carrier, the killing or inhibiting effect of the cytocoxic substance be e~phasized or amplif ed.
Thus, the therapy achieved by the method of claim 1 is an important step toward this objective.

132330, According to one aspect of the invention there is pro~ided the use in the treatment of malignant diseases a covalent conjugate medicament consisting essentially of the following components bound covalently together, a) a cytotoxic substance, b) a poly~.er carrier having an endocytic biodegradable, nontoxic, polyaminoacid backbone, c) a cell homing vector with the properties of first selectively targetting the conjugate toward malignant cells to be fought and second, providing for the internalization of the conjugate into said cells, wherein fast biodegradation of the carrier will occur with consecutive release of the cytotoxic drug which will selectively combat said malignant cells.

According to another aspect of the invention there is provided a cytotoxic targetted conjugate for the application of the method of Claim 1, this conjugate having the formula Z
- ~G~ Cv -F- ~ - H-~

(lc~;2~p ~rly ~0 ~_ CO-.~. ~-DIY CO-.~. ' y~`_~!' in which p and o are 1 or 2;A and A' represent chain extending amino-acid intermediate links of formula -~NH CHY-C0) and x and y, which define the number of these links per molecule, can be zero or any integer from 1 to 20; y is an amino-acid rest; DM and DM' represent one or more cytotoxic substances covalently bound to the aminoacid carboxyl group through an _,.

1 3 2 3 3 0 ~

amide or ester lin~ but DM can also represent an OH of a free carboxyl; EGF defines a homing vector for promoting malignant cell recognition and internalization therein; R is a group of formula.

-C~ a- ( G~ iH wi.h ~ ~einq ~2- ~r~ e~

~rom -(C~ with r .r ~ d; or ~ ; o~ ~ ; ~

In some embodiments, the factor promoting cell internalization of the homing vector (c) also acts as the "arrow-head", i.e. it recognizes and binds to the cells at suitable receptors thereof. Examples of such vectors are the epidermal growth factor tEGF) or other EGF analogues of growth factors such as urogastrone, -TGF, or synthetic and natural derivatives thereof, transferin, low density lipoprotein (LDL),Nerve Growth Factor (NGF), Platelet Derived Growth Factor (PDGF), and some viral receptors, all providing cell internalization.

~O Otherwise, in some other embodiments, the homing vector can comprise separate binding and internalizing factors; for instance, the binding element can be non-internalizing antibody (such as FAB) to a specific antigen of the cells and the internalizing element can be any known internalizing factor specific or unspecific.

The use of EGF factors to specifically direct medicaments toward EGF receptors on malignant cells is not unobvious per se since several investigations have demonstrated the -3a-. ~ ~

1323'~'~

presence of these receptors in high concentration on many squamous cell carninomas as well as sarcomas. Such results include breast, lung, brain and skin tumors (see the following references; J. Hunts et al., (1985) JPn. J. Cancer Res. 76, 663-66; B. GUSTERSON et al., (1985), The Lancet, 364-68; F.J. Hendler et al., (1984), Cancer Res. 443, 753-60;
T. BAUKNECHT et al., (1985), Dermatologica 171, 16-20.

It is known also that these receptors are particularly efficient targets for EGF-linked cytotoxic drugs because of internalization into the -3b-13233~ J
cells upon interaction with the EGF-receptors (see for instance T.H. HAIGLER et al., (1979), The Journal of Cell Biology 81, 382-394). Exemplifying the targeting of a drug with EGF is provided by document W0-85/03357 (MD. WATERFIELD) which discloses antibodies recognizing EGF receptors which effectively inhibit the activity of these growth factor receptors in tumor cells.
Another example is provided by an article of N. SHIMIZU
et al., in FEBS letters 118 (1980), 274-278, who reports the preparation of a conjugate between EGF and diphtheria toxin.
However, as far as the present inventors are aware, no therapeutic method using a conjugate of EGF (or analogue) with an endocellular drug-release polyaminoacid back-bone carrying a cytotoxic su~stance in high mole-ratio and with a so impredicatably potent effect (as will be seen hereafter) has been reported.
The new therapeutic compounds of the present invention using EGF or analogues as a homing vector can be illustrated by the formula below:
~--~
EGF-R-(C0-~H-NH)-~..- Cv-CH-NH)~n~~

(I 2)P (I 2)o (I) - C0-A~-DM ~A~y~~M~
in which EGF designates the homing vector ~c) as previously defined.
In the above formula, p and 0 are the integers 1 or 2; A
and A' represent chain extending amino-acid links of general formula -(NH-CHY-C0)- and x and y define the number of these links. Actually x and y can be zero or an integer from 1 to about 20. It should be made clear that A and A' can define a sequence of identical or different amino-acids, i.e. in each X

~3233& ~, link Y can be the same or can be different. Y defines the rest of the amino-acids in question; for instance Y can be H
(glycine), CH3 (alanine); iso-Bu (leucine), benzyl (phenylalanine); p-hydroxy-benzyl (Tyrosine), -CH3-COOH
(aspartic acid); -(CH2)-COOH (glutamic acid), and other rests of other aminoacids. The nature and the length of the A and A' extenders allows to 4a ;F

13233Q, control the biodegradation rate of comp.und I ar.d '~enre the re'ea~o ~f the la~el ~M. In genæral the strucrllrP of the pol~mer ~ack-Done and the chain extending segmen.s is adapted for qul~-k biov~ograrion b-~ the endsce -lular enzyma.ic system. For instance a polyglutamic structure i3 readil-~deyraded by endocellular a-glutamylt.ansferase ~i~h fast release of the cytotoxic label. hhen re~uired the aminoacid st ucture of the polymer bac.~-bone and segments ~ and A' can be adap~e-~ for preferential attack by ot.~er endocellular en~ymes.
Ir. formula I .'1~ and DM' represer.t the sami or different cyt5~xic substances selected from Dauno~ycin Ac~iam~cin me ~otre-.ate ar.d any of the cytotoxic substances recited in the -ntroductior. and t.'e ~rior ar .
Ncrmaliy the cytotoxic substance is Do~nc (by m.e~ns o~ a ?r~ary a~i?e function thereof) through an amide linc to the terminal carbo~y group.
Since the e~;ten- or bir.dir.g of the cytotoxic substances i3 general'y belcw lOQ~ scme of the carboxy'ic end-group in rormula ~ are free anc p2rr of the DM or DM' groups represent ar. -0~ Jroup. A .on~enien.t wa-~ of ?ict~ring this situation wlth formula I is to cefine ~M as the c-ytotoxic substances DM' as an -OH and the ratio m/n as the deg~ee of amidation (or esteri ica-tion! of the carboxylic end-grou? 3y the cyto-oxic subst~nces. .hus the carbcxy'vte group in compound I is there for er.sur ng -~a'er so'u~ y. The ind ces m arc n rerpresent numbers 3uf.-'c.en; -o ?ro-~iide mole.1'ar ~eights !~r about 50CO ~o 100~00 ~a ar.d n/r~ is in ~ e ance 2 to 'O.
In ror~.ula I ~ is a group of fcrmula IVI) i.e -CO-X-N ~ -S ~ ~H- where ~ ~s a bridg-ng o hyZrocarbon chaln selected from alky_ene (~ith 1 tO 4 CH2!, 1, 3-cyclohe-gylene m-phenylene p-toluylene p-trime.hyler.e-phenylene e'c.
In for~ula VI the acyclic carbonyl can aiso be a thiocarbonyl.
As said before svmbols A and A' represent si?.gie aminoacid units or oligo?ep.-dês _~ntaining ~rom 2 to 70 a.~~noa- d U?i's uch o'igo?e?tides are acapred tv ?ro~tide control'ed h~drol~r'~ (en~-~ar'-) re'ea_e o~ the drug dependins on the selected target s te. mhe am-no aclds selected for m2king the A ard A bridges can be any usual am~no-aclv li~e alanine glycine leucine serine pheny;alarine arglnine et.... a"d the-y vare sele_~ed in n~mbver anc kind depencilng cn .he needs ~nà .h- a??:i-ation.

132~ .~l` i This technique of making the release of the drug controllable by linker structure has been detailed in applications EP-A-150,184; EP-A-130,935 and WO-PCT-CH86/00177.
Compound I can be prepared by coupling a 4-(N-maleimido) derivative II of EGF (or other EGF activated compound) with the thiol-copolymer III according to the following scheme (in this scheme Z designates the polyglutamic moiety of compound I) Q,_ EGF-NHC0~ H~ N-Z ---~ I

Compound II, can be prepared by the activation of EGF
with N-succinimidyl-4-(N-maleinimido)-butyrate, but other heterobifunctional bridging compounds (see below) can also be used to activate EGF and can be prepared according to the aforementioned KATO reference. Said activation consists in reacting one terminal -NH2 of the antibody on the butyrate carbonyl, thus providing the -NHCO-link in formula II.
Other agents suitable for activating EGF, thus providing compounds usable in place of II in the above scheme, are those having at one end a reactive NHS-ester functional group and a sulfhydryl reactive group at the other end; exampIes of such compounds are the m-maleimidobenzoyl-N-hydroxy-succinimide ester, m-maleimidobenzoylsulfosuccinimide ester, succinimidyl-4-(N-maleimidomethyl) cyclohexane-l-carboxylate, succinimidyl-4-carboxylate, succinimidyl-4-~p-maleimidophenyl) butyrate, sulfosuccinimidyl-4-~N-maleimidomethyl) cyclohexane-1-carboxylate, sulfosuccinimidyl-4-(p-maleimidophenyl) butyrate, etc. A
good description of the use of these bridging agents can be found in the PIERCE 1985-1986 Handbook ~ General catalog, page 326 onward; other known suitable bifunctional compounds could also be used.

~r~

i.3233Q .~, Labelling the intermediate polyaminoacid backbone of the intermediate polyaminoacid-thiol (i.e. formula III in which DM and DM' are still OH) with Daunomycin, or any other cytocide, can be done also according to KATO, J. Medical Chem. (1984), 1602-7 by first protecting the thiol function by a 2-pyridyl-disulfide group and reacting the protected intermediate V in the presence of 1-ethyl-3-(3-(dimethylamino)-propyl) carbodiimide hydrochloride (EDCI) according to the following scheme, then removing the protective thiol-pyridine with dithiothreitol (DTT).
ITI ~ ~ -SS~ S~ ~!iH-Z

(DM in Z is 0~ M in Z is OH) V

~ -S~ ~ ~H-Z + DM ~ IlI
~ 2. DT. (r~ = Qbuno~ycin) EGF, a single 53 aminoacids polypeptide, can be extracted from submaxillary`glands of mice (see J. SAVAGE et al., ~1972) J. Biol. Chem. 247, 7609) or it can be synthesised by genetic engineering methods, for example using a cloned gene. EGF can also be of human origin.
To introduce the chain extension segments A and A', usual techniques prevalent in polypeptide chemistry can be used. Such techniques are detailed for instance in the foregoing references EP-A-130,935 and WO/PCT-CH86/00177.
Compound I has other advantages in addition to being t internalized into malignant cells and controllably releasing therein its bound cytotoxic drug. Quite unexpectedly, its selectivity toward malignant cells over that of previously known cytotoxic drugs is considerable as will be seen hereafter. Further, the polymer construction should prevent 132~3Q ~
the polymer drug complex from penetrating into heart tissue, thus removing one of the major shortcomings of free daunomycin and other known antitumoral drugs.
Reference is now made to the accompanying drawings in which:
Fig. 1a is a photomicrograph of a culture of A-431 human squamous carcinoma cell line used to test the effect of the compound of the invention. The presence of large amounts of EGF receptors thereon is indicated by indirect immunoperoxidase staining (dark color).
Fig. lb is a photomicrograph similar to that of fig. la but of a culture of normal WI 38 embryonic fibroblast cells used as a control. The low level of EGF receptors is indicated by the quasi absence of staining 7a ~ i _ 13233G~
(light color).
Fig 2a is a photomicrograph of an A-431 cell line culture after a 4a hrs incubation with 1 yg/ml of free daunomycin and testing for dead cells with Trypan Blue exclusion dye.
Fig 2b is a photomicrograph similar to that of fig. 2a but after 48 hrs incubation Nith, instead, 1 ~g/ml of Dbunomycin in the form of compound I. The dark areas represent the killed cells.
Eig. 3a is a photomicrograph at time zero of a mixed culture of A-431 and r~I ~8 cell lines. The round cells are the tumor cells.
Eig. 3b is a photomicrograph si~ilar to that of fig. 3a showing the situation after 24hrs incubation with 1 ~g/ml of compounZ I. the round tumor ceils have greatly diminished.
~ ompound I demonstrates very useful properties in cancer therapy. A
first valuable property relates to iis strons affinity toward malignant cells over normal cells. Eor demonstrating this characteristic, a human squamous carcinoma cell line was selected, for instance A-431 cell line shown in fig. la (see M.~. WATERFIEL~ 8,), J. Cell. Biochem. 20. 14~-161), although other tumor cells can be used as well. The membrane of these cells contains a very large concentration of EGF receptors which makes them highly suitable for test wi'h compound I. The presence of these receptors is shown by indirec' im.munoperoxidase staining and appears as dark color areas of fig la. Control cell lines, for instance WI 38 embryonic fibro-blast cells with low amount of EGF receptors ~see fig. lb), Nere treated identically for comparison and appear as lighteer color areas on the photo-micrograph.
- Thus, when a known amount of compound I (p = o = 2; x = y = O, DM =
daunomycin; DM' = OH; n/m = 6) radio-labelled with I125 ~iodine is part of the EGF moiety) was incubated with A-431 cells using WI 38 cells 2S con-trol, 31% of total radioactive iodine was retained in the intra-cellular compartment and 2% in the membrane of the tumor cells, ~hile in the cGntrol cells, the corresponding values were 2% and 0~, respectively. Using free EGF in place of I in these experiments gave the corresponding 14%/0% and 7%/0% values instead. The reason of the greater accumulation of compound I
as compared to free EGF is possibly due to retention of EGF by the polymer backbone of compound I by endocytic vesicles, whereas EGF is recycled freely back to the medium.
A second valuable and unexpected ?roperty of compound I relates to its enhanced toxicity as compared with free daunomycln toward malignant cells.

Thi~ is illustr3ted by tigs 2a and 2b.
In flg ~a, d cui~ure of A-431 cells is shoNn after 4~ hr_ incubation with a med~u.~ contal`nlng `~ ug/ml or free daunomycin. ~/iability ~as scored by ~eans of the Trypan ~lue exclusion dye ~ethod. E~clusion of the dye from the cells demonstrates the viability thereof. In contrast, when an equiva-lent amount of daunonycin in the form of compound I was used in a s~ilar experinent ~see fig. ~b), a very large number of ce'13 were :~-lled as shown by the shrivelied up cells and the dar~ areas where the dye 'na~ accumula-ted.
The seiectivity of compound I to~ard tumor celis was further tested.
For instance, fig. 3a and 3b show the effect of compound I in the case of a mi~ed culture of A-431 andd WI 38 cells. The round celis repre~ent the A-431 tumor celis ,~nd the elonsated cells are the WI 3~ control cells. The culture medium was Dhot3sraphed at time -ero t'ig. 3a). Eig 3~ illustrates this situa-ion after ~4hrs in a medium conta-'nir.g i ~g/.~.l or compound I. It can be seen tha~ t~e round darker malignant cells have stron~ly re~ressed.
In conclusion, it has been demonstrated that at equiv~ient molar concentrations (of daunomycin) com?ound I rAas much more lethal to squamous carcinoma ce'ls than free daunomycin itself. Also compound I has a selec-~ive mortal ac~ivity on tuncr cells ~ut leaves normal cells allve, ~hereas, under the same cond '._ons, free daunomycir will kil' norma' cells.
Further advan-ages of the compour.ds of the inver.t1cn ror treatlr.g-tu~or diseases relate to the follo~inc characteristlcs:
Molecular size: The compounds or tne invention are smaller than most usual targetted drugs using monoclonal antibodies, wherefrQm improved pene-tration into tumor bodies is assured ~ith consequent better cytoto~ic efficiency. Yet, the compounds Ot the invention are bigger than the free cytotoxlc drugs ~hich ?revents penerr3tion lnto nornal cells. Ir contrast, malignant cells being susceptible to endocytocls they will allow penetra-tion of compounds of moderate size like that of the invention.
Effect of EGF and related factors: EGF has an an~iogenic effect and develops mali~nant cell vascularization wi.~ consequent cell mitosis and increased suscepti~ility to antimitotic drugs. In otner words, t~e com-?ounds of the irventicn tenporarily incroase the ~umor cel' metabolism ar.d render it more sensitive to d.ug killing actior.
The following Exam?les lllustrate the invention in more detail.

1~2~30' EXPERIMENTAL SECTION

Example l Synthesis of compound I, a conjugate of EGF and daunomycin-grafted polyglutamic acid.

A polymeric compound ~V) with a poly~lutamate ba-.'ibone structure (p o = 2; 3 = y = O, in Nhich some ~ -carboxylic groups are condensed with daunomycin and having a 2-pyridyl-dithio-ethylamido heaZing group was pre-pared according to Y. KATO et al., (lg84~, J. Med. Chem. 27, 1602-1607 (compound 5 in scheme II of KATO).
The identity of the compound was chec.~ed analytically:
Mw = 29000 Da by quantitatively determining the 2-pyridyldithio group;
ratio; ratio of daunomycin to carboxylate = 1i6 as determined by spectrome-trlc quantitation of daunomycin at 480 nm.
To twenty five mg of the polymer dissolved in 2 ml of lOmM sodium phosphate buffer at pH 7 were added O,l ml of a 0,3 M solution of dithio-threitol (~TT). After l hr at 40C the solution was dialyzed overnight against a 0,1 M sodium phosphate buffer at pH o,O (SPECTRAPOR membrane, MW
cutoff 3500); this regenerated 'he thioethylamido group of the molecule (compound 6 in scheme II of ~ATO); yield ~3 mg of a red compound after free2e-drying (poly-(DM)-Glu-SH). The poly-(~M)-Glu-SH polymer was reacted with an excess of thiopropyl-SEPHAROSE~in the pyridylsulfide form (4C; 12 hrs; phosphate buffer, pH~) and the gel was rinsed with an excess of the same uffer in order to eliminate the polymer lackin the SH extremity. The polymer was conserved ur.der this form in the cold. It was then regenerated by treating the gel with an excess of mercaptoethanol (12 hrs), dialysed against water (overnight, 4C) and freeze-dried before reacting with EGF.
One hundred ~g of EGF (SIGMA) ~as dissolved in 500 ~1 of lO mM phos-phate buffer, pH 7,0 containing 0,14 M NaCl. Then a quantity of l25I-EOE
sufficient to provide an activity of lO,OOO cpm/~g EGF was added followed by 50 ~1 of a 32 ~ solution of N-succinimidyl-4-~-maleimido)-butyrate (SMBU) (origin: Sigma), in dimethylformamide. The mixture ~as allo~ed to stand for l hr at 25~C, then it was dialyzed aga~nst a O,l M sodium phos-phate buffer-O,l M NaCl,, pH 6,0, to eliminate the excess of SMBU.
The desired product (II) resulting from t~e cor.densation of S~BU and EGF was not isolated ~u to tne 500 al of the d aly-ed ~GF solution ~ere 132330~

added 1 mg ot poly-(DM)-Glu-SH and the mixture ~as slowly agitated over-night at 4C. ~ive ml of thiopropylsepharose (pyridylsulfide form) were then added and the reaction was continued at 4C for 12 hrs. The gel was washed with successively 3 portions of l ml of sodium phosphate buffer, the eluent was concentrated under reduced pressure and subjected to gel filtra-tion on SEPHADEX G75~ column 40 x 0,8 cm), using 0,l M ammonium carbonate solution pH 7,0. The fraction containing the EGF-poly-(~M)-Glu conjugate ;I), absorption 490 nm, was collected and freeze dried. Yield: 80 ~g of solid.

Example 2 Selection of A-431 test cells lines from evidence of receptor concentration on the cell membrane by indirect immunoperoxydase stain-ing Indirect immunoperoxydase staining on cell lines were performed on trypsinised cells in 35 mm P~C plates. The surface was pre-treated with phosphate buffered saline (PBS) ph 7,~, the excess was then removed and the PBS washed cells (105/well in 50 ~l ?BS) were added to the plated and centrifuged for 5 minutes at `2000 rpm. 50 ~l/well of 0,5% glutaraidehyde in cold PBS were then added to the dish and incubated for 15 minutes at room temperature. After two round of washes with PBS, the wells were filled with 100 mM glycine in a 0,1% BSA solution and allowed to rest for 30 minutes at room temperature to block gutaraldehyde activity. After two PBS washes, indirect immunoperoxydase was done by first denaturing the cells with an ice cold mixture of 9~:l ethanol-acetic acid for 30 minutes at 4C. During this period, 3 ~1 of antibody were diluted in 1 ml PBS + 1% foetal calf serum (FCS). The wells were then washed twice with PBS and incubated for 5 minutes with a solution of 20~o FCS in PBS. This was then replaced by 200 ~l/well of the antibody solution and incubated for 30 minutes at room temperature. The wells were then washed twice with PBS, once with PBS +
Tween 0~l~o and once again with PBS. 200 ul/well of a 11400 dilution of swine anti-mouse perodydase ~POD) conjugated ant.body (DAK0) in PBS l~ FCS
was then washed twice with PBS, once with PBS + 0,1% tween and twice with dlstilled water. The in situ coloring was achieved by incubating the cells at room temperature with a solution of lO ml 0,Ol M phosphate buffer pH
6,0, 5 ~l 35~O oxygenated wa~er and lQu ~l of l~, ortho-àianisidi.. (MERCC) in methanol. 132330~

Example 3 Internalisation of compound I in A-431 squamous carcinoma cells compared to in HI-38 fibroblasts The entry of compound I into the cells is essential for .t3 action.
Therefore, internalisation Ot the molecule was demonstrated by showing its presence in the membrane and inside the cell compartment. This has been done using A-431 and WI-38 cells. t Compound I, radiolabeiled with 125I on the EGF, was incubated with confluent cell cultures for 6 hrs at 37C in solution A (4 parts ~MEM and 1 part of 50 mM Tris, 1~0 ~M NaCl and 0,1% BSA adjusted a. ph 7,4). The cells were then washed ~our times ~ith ice-cold PB~T (lmM Cd Cl2and 1 .~M
MgC12). Fifty % trichloroacetic acid ~as added in a proportion of 1!5 to the pooled solution a and PBS+ and counted on a y counter. The cell mem-brane was destabilized by a treatment on ice with 200 mM acetic acid and 150 mM NaC1 (solution B) for 6 minutes. The solution B was then removed and the cells wa~hed tNice with solutlon B. These pooled solutions were assayed for 125I. This treatment releases the EGF rece?tors bound to the cell surface. The cells were then complete1y dissolved in 0,7 N NaOH. The radioactivity found then represented the internali,~ed compound I. Ln con-trol experiments, using free EGF 125I, a proportlon of the EGF is recycled to the medium therefore lowering the intracellular EGF. The results on the internalization of 125I labelled EGF and compound I in ~1-38 and ~-431 cell cultures are given in the following Table in terms of counts per min (background 30 cpm) for successively: EGF not integrated in the cell, EGF
bound to the membrane, and EGF in the intra-cellular compartment. The percent of total cpm is given in brackets.

132330 ~
TABLE

EGF (%) Compound I (%) WI-38 530 (93) 1169 (98) " 29 (O) 28 (O) " 67 (7) 52 (21 A-431 536 (86) 831 (67) " 31 (O) 57 (2) " 111 (14) 390 (31) These results show that compound I is more efficiently internalized in malignant cells (31%) than in normal cells (2%~.

Example 4 Comparative cytotoxic effects of free daunomycin and compound (I) on a A-431 squamous carcinoma cells All cells were maintained in Dubelcco's Modified Medium (DMEM), 10%
foetal calf serum (FSC) (Gibco), 2% penicillin-streptomycin (Gibco) and 1%
fungizon (Gibco~ in 5% C02. They were plated at 50 to 60% confluency 24 hours before the addition of the toxin. 1 ~g/ml of daunomycin, or equiva-lent in compound I, was added in DMEM 10% FCS and the ceil death rate visualized by trypan blue exclusion. 4 volumes of 0,2% (w/v) Trypan Blue in water was freshly mixed with 1 volume of a saline solution 4,25% (w~v) of NaCl in water. 1 volume of this solution was mixed with 1 volume of PBS on the cell monolayer or to 1 volume of cell suspension in PBS. Observation and scoring took place 48 hours after addition of the toxin (see fig. 2a and Zb). These results show that daunomycin is much more effective against malignant cells when in the form of compound I than when in the free state.

14 13233~

Example 5 Effect of compound I on mixed cultures of tumor (A-431) and normal (~I-38) cells.

Cells were maintained as described in e~ample 4. The ~I-38 fibroblasts were first plated and the A-431 squamous carcinoma cells were plated the next day. The mixed culture was left growing for 24 hours and then 1 ,ug/ml of compound I was added to DMEM 10% FCS. Nearly all the A-431 cells were selectively killed after 24 to 48 hours whereas the ~I-38 fibroblast were left allve. Hence, the mixed culture became free from the tumor cells demonstrating that compound I can be used to separate the normal cells from cancer cells. The experiment was repeated but using only 0,1 ~g/ml of compound I (for the controls, equivalents of daunomycin were used in free form). Observation of the cultures after 15 days showed that the test samples contained no more of A-431 cells, which situation was confirmed by continuing culturing for 6 weeks under normal conditions, this resulting in no reformation of tumor cells. In contrast, in the controls all cells, malignant and normal, had died after 15 days.

Example 6 A compound of general formula V (see description) was prepared accor-ding to KATO et al., G. Med. Chem. 27 (1984), 160Z-1607 (compound defined as 4 in scheme I of KATO). This compound has the formula VA below in this paticular embodiment.

SS ~ NH~(CO~CH~NH)q~~H VA

( C;~2 ) 2-COoH

in which q may be fro~ about 150 to 250 depending on the polymeri7ation conditions.
This compound was subjected to a procedure reported by HESS-~IJK e~
al., J. of Controlled 2elease 1 (4) (1985), 312 for e~tending the side chain with an H-Gly-Gly-Leu-OH peptide (segment A in formula I), as fol-lows: 40 mg of VA and 68 mg of saccharin (0.31 mmole) were dissolved in 1 ml of DMF and the solution was allowed to rest far a few hours (solution A).
On the other hand, 0.32 mmole (40 ~1) of N,.~,N'~'-tetr~methylganidine (TMG) ware slowly added to a stirred suspension of 0.3Z mmole (8C mg) of H-Gly-~ly-Leu-OH in DWF Stirring was continued until all sollds had dissolved ~solution B).
Then 0,43 mmole (70,3 mg) of ~,N'-carbonyl-diimidazole were added to soiution ~ and, after stlrring for 30 min, solut~on 3 waa added. The mi~ture was further stirred for 3 days at room-tempera~ure. The mi~.ure was added into 15 ml of 0.1 M phospha~e buffer (pH 7.0) ar.d the resulting solution was dialyzed into water (17 hrs) filtered on a millipore membrane (0.45 ~m) and the filtrate was freeze-dried. The polymer ~yield 80% mg) was analy~ed by hydroly~ing an aliquot in ~ N HCl for lZ nrs at 180C. Deter i-nation of the aminoacids in the hydrolyzate was carr_ed out ~y High Perfor-mance ~iquid Chromatosraphy of the amlnoacld-~Atalaldeh~fde derivatives (detection by fluorescence). The followlng ratlo of glutam~c~lycine/'eucine was measured: 0.95/2/i. These results indicate that the grafting level was aroung 95%r i.e. the compound can be represented conventionally by the formula:

PYR-SS ~ ~lH ~ CO-jH-NH)- 05........... -(C0-jn-NH) ~5 ~ . H

(CH2)2 COO C~.2(CH2CONH)3-CH(iso'Bu)COO

as the tetramethylguanidlne salt. ('~B) The pyrldine-S group was removed ~ith dithlothreitol according to KATO
et 21., and analysis Nas performed by measuring the absorbance at 343 nm of the ll~erated pyridine-2-thione. Neqlecting the presence of the TMG+ ion, a molecular weight of 25.500 was found meaning that in the product t (the degree of polymerizat on) i3 about 75-90.
Labellinq compound ~B with Daunomycin waa ac_om?liahed ~s follows:
33.7 mg (74.7 ~mole) of '~B ~ere disolved in 15 ml of ~ a(iueous NaCl and 20 mg (35,5 ~mole) of 2aunomycin hydrochloride were added. The pH ~as brought to 5.5 Nith 0.1 N ~aOH arter which ~8 mg ~0.1 .~r~cle! of 1-ethyl-3-C3-(dimethylamino)-carbodiimide hydrochlor~de ~EDC) ~ere added under s;irring.

16 1323"\J~

After agitation for 18 hrs, the mixture was dilute~ with 15 ml of 1 M NaCl and dlaly7ed in ~ater~ The residue was free7e-dried wh ch provided 31.5 mg of polymer. Splitting t;r~e dlsulfide with DTT as before and analy7ing spec-troscopically the pyridine-thione indicated the prosence of about 10 DM per molecule, i.e. a ratio of labelled side-chains to unlabelled side-chains of about 1:7.
Conversion of an aliquot o~ the above disulfide to the deslred cor-res?Gndi.rig thiol I -~B with DTT was done as Ec~ ows: 2 mg of pol-~me- were dissolved in 2 ml or 0.1 ~ phosphat- buEtar (?H 7.0) and 150 ul of 0.3 M
DTT were added. After allowins tG stand for one hr at 42, the solution was dialy~ed for 4 hrs agaln~t freshly degassed phosphate buffe. ~pu 6.0); A
SPECTRAPOR bag was used (MN cut-off = lOOC).
Simultaneously, EGE was ai-t~ated by tak ng 0,2 ~g of _GE (Sigma) and dissolving in 3.?5 ~1 oE 10 .~M ?hosphate bu~ter in 0.14 M NaCl, pH 7.0;
the-, adding 0.5 ml of 2 10 ~g/ml S~BU solur on in DI~F. After 2 hrs at '0C, the mi~ture was dialy7i?d at d~C ~gainst a 0. ~ Phosphate ~uffer contalning 0.1 M NaCl at pH 5.0 'membrane Mw cut-oEf = 1000).
The overall wlume of both the dlalyzed polymer and dialy~ed EGF
solution Nere reduced to 1 ml by absorption with C`~C powder, then they were mixed together and allowed to stand for Z4 hrs n one dialysis Jas. The m :~ture was c;lrcmatGsraphed on S~P~ADE~ G75 ~eluent O.lM N~o~ and the fract ons absorbing a~ 4~0 nm were collected and cleaned from unreacted ?olymer by treating with thiopropy sepharose overnight at 4C. The gel -~as washed with O.lM NH4Co3 and 22.5 ml (O.D. of 0.176) of solution was collected. Yield about 80% of compound IA. The respective weight contri-butions of DM and EGF in the product are about equal.
If, in the above pre~aration, Daunomicin is replaced by Adriamycin, a similar product, incorporatins Adriamycin is obta ned.
The presence and bir.dlng efficiency of the EGF factor in .ompour.d IA
was checked by immunoprecipitation with EGF antibody andd attachment of the immunocomple~ to a protein A - Sepnarose gel (C~4B, Pharmacia). The proce-dure was as follows:
Protein A - Sepharose gel was rahydrated to ~rovide a 50~, (VtV) solution in NET-NP40 buf-er (100 .~M NaCl, 1 .~ A, 10 ml`~ Tri,, pY 7.5, 0,5% NP40). Bovine serum albumin (BSA) was added to 20 ~1 or the buf ered sepharose solution to provide a 0,3% (by weight~ ~SA solution ~S!. On the o~her har.d, BSA was added to 12 ul of a sc uticn of antiserum aga-nst EGF
(Collaborative ~esearch) so as to ?rc~; d- ~ 0,_~ by weiqn. BSA so u _on in 17 13233G, antiserum (Ab). ~oth (S) and (Ab) were incubated overnight at 4~C, then a quantity of compound IA corresponding to 120 ~g of daunomycin was added to sample Ab and incubated for 7 hrs at ~C under agitation. Then solution (S) was added and the mixture was agitated per 12 hrs at 4C. A control (C) was prepared by adding the same quantity of compound IA to another identical sample of solution (S). Both the above mixture (M) and the control were centrifugated for 5 min at 2000/rpm and the optical density (OD) of the supernatant liquid measured at 480 ~m. The results are 0.41 for M and 0.955 for C which shows that compound IA c~rries EGF, the conformation of which is recognized by the ~ntibody on the gel.

Example 7 The toxicity oE compound IA and of free daunomycin toward A431 malig-nant and W~3~ (control) cells was compared. The effect of these drugs was evaluated by the degree of inhibition of cellular protein synthesis. For this, we measured the level of incorporation of 35S methionine (35S-met NEN) into newly synthesized proteins after a 48 hrs exposure to different concentrations of the drugs.
Cells were plated on 1,5 cm Petri dishes at 50% to 60% of confluency before the addition of the toxin. 0,1, 0,5 or 1 ~g/ml doses of daunomycin (controls) or its equivalents in the form of compound IA were added in DMEM
10% FC~. Cell death consecutive to this addition was measured as follows:
The cells were exposed for 1 hour at 37C in 500 ~1 DMEM low methionine medium (Gibco) containing radioactive 35S met. The medium was then removed and the dishes were washed with PBS (137 mM NaC1, 2,7 mM KU , 8,1 mM
Na2HpC4, 1,5 mM KH2P04, 0,9 mM CaC12, 0,5 ~M MgC12 pH 7.2). These solutions were stored for ~ counting. The cells were lyzed with 1 ml 0,1 N NaOH and placed in a tube Nith 500 ~1 Trichloroacetic acid (TCA) 10% to precipitate the proteins. This mixture was filtered on GF/A filters (Whatman) to eliminate unreacted 35S-met. The filters were washed twice with 1 ml of 10%
TCA and once with 100% Ethanol. The filters were dried at 80% for two hours and sub~ected to ~ counting in Econofluor (NE~) sc-ntillation medium. The results reported in the following table show that compound IA is very cytospecific but less cytotoxic than daunomycin alone.

18 ~233aj drug concentration % Oe Protein synthesis inh~bition (~g/ml A431 WI38 0,1 30% 10%

FREE DAUNOMYCIN O,5 69% 46%

1,0 97% 80%
=========================================================
0,1 4% 0%

CONPOUND IA O,5 17% 6%

1,0 47% 4%

Example 8 IN VIVO TEST OF COMPOUND I

We induced A431 xenografts in nude mice and when the tumors reached 3 to 5 mm, we in~ected into two sets of nude mice 0,1 mg/kg of daunomycin either in the form of compound I (see example 1) or in free form. In the prior art, 10 mg/kg doses have been used in mice (Schwarz S. et al., 1975, Cancer Chem. Rep., 6, no 2, 107-114). The drug was injected either directly in the tumor or in the caudal vein 4 times at 3 days intervals. The measu-res were taken a week after the last injection.
By visual inspection of the animals, it was noted that the growth of the tumors treated with compound I was significantly reduced, as compared to control mice or to treatments with free daunomycin. These results were similar whether the injection of the drug was directly in the tumor or in the caudal vein which shows that biodegradation and release of the cytotoxic substance occurs substantiallly only in the target cells. Results are shown in the follo~ing Table (I.V. = intra-veinous; I.T. = intra-tumor) 19 1 3 2 3 c~ Q

injection initial surface final surface growth of the tumor ~A) of the tumor (8) coefficient (B/A

Daunomycin I.V. 30.5 500 16 Compound I I.~. 25 54 2 Daunomycin I.T. 9 170 18.7 Compound I I.T. 9 50 5.6 Control* 9 320 35.6 * Placebo injection At the end of the experiment, the tumors were dissected and weighed.
Results are given below. If corrections for slight differences of tumor si~e at the beginning of the treatment are taken into account, it can be seen that the results correlate well with the surface estimate glven in the previous Table.

~aunomycin I.V. : 1.178 g.
Compound I I.V. : 0.359 g.
Daunomycin I.T. : 6.789 g.
Compound I I.T. : 0.816 g.

in conclusion, the compounds of the invention show better performances in selectively killing squamous carcinoma cells than daunomycin, both in vitro and in vivo. In the animal tests, we noted that very low amounts of com-pound I have a remarkable effect on tumor growth.

E~ample g The in-vivo tests reported in Example 8 were repeated using compound IA instead of compound I. Thus, 2 mg/kg of daunomycin, free or in the form of compound IA were injected every 3 days over 9 days in the caudal vein of nude .mice bearing A431 tumors. The tumor growth inhibitlon by compound IA

132330.~

was significantly greater than by free daunomy.cin (D), free EGF or DM
labelled but untargetted polymer (DMA). These results are shown in the Table below where the values correspond to tumor diameter (in mm) measured with a scalliper after a number of days. The lethal dose of compound I and compound IA have not been measured but are presumably less toxic than free daunomycin which enters freely into most normal cells.

T A B L E

Dayg 0 4 7 11 14 Control 10 14 17 2Z 24 Free EGF 10 13 14 17 19 DMA 10 11 12 16 i7 IA 10 11 12 14 i5

Claims (16)

1. The use in the treatment of malignant diseases, of a covalent conjugate medicament consisting essencially of the following components bound covalently together, a) a cytotoxic substance, b) a polymer carrier having an endocytic biodegradable, non-toxic, polyaminoacid backbone, c) a cell homing vector with the properties of first selectively targetting the conjugate toward malignant cells to be fought and second, providing for the internalization of the conjugate into said celis, wherein fast biodegradation of the carrier will occur with consecutive release of the cytotoxic drug which will selectively combat said malignant cells.
2. The use according to Claim 1, wherein the homing vector also comprises one or more EGF, EGF analogues such as urogastrone, ? -TGF, and synthetic or natural derivatives of growth factors providing cell internalization as well as tumor cells specific markers.
3. The use according to Claim 2, wherein the homing vector also comprises one or more antibodies capable of selectively bind to a particular antigen possessed by a malignant cell.
4. The use according to Claim 1, wherein the polymer carrier is polyglutamic or polyaspartic acid.
5. The use according to Claim 3, wherein the polymer carrier is a copolymer of aspartic and/or glutamic acid with other aminoacids such as glycine, leucine, valine, isoleucine, alanine, phenylalanine.
6. A cytotoxic targetted conjugate for the application of the method of Claim 1, this conjugate having the formula (I) in which p and o are 1 or 2; A and A' represent chain extending amino-acid intermediate links of formula -(NH-CHY-CO) and x and y, which define the number of these links per molecule, can be zero or any integer from 1 to 20; Y is an amino-acid rest; DM and DM' represent one or more cytotoxic substances covalently bound to the aminoacid carboxyl group through an amide or ester link but DM can also represent an OH of a free carboxyl; EGF defines a homing vector for promoting malignant cell recognition and internalization therein; R is a group of formula.

with x being selected from -(CH2)r with r from 1 to 4; or ; or ; or , or ; m and n are integers of value sufficient to provide a molecular weight of 10,000 to 500,000 Da, the value of m/n being 1/10 -1/2.
7. The cytotoxic conjugate of Claim 6, in which said homing vector comprises the epidermal growth factor (EGF), EGF
analogues such as urogastrone, .alpha.-TGF, and synthetic or natural derivatives of growth factors providing cell internalization as well as tumor cells specific markers.
8. The cytotoxic conjugate of Claim 6, in which the homing vector further comprises one or more antibodies capable of selectively bind to one or more particularly antigens of malignant cells.
9. The cytotoxic drug of Claim 6, wherein DM and DM' are selected from auromycin, hematoporphyrin, platinum complexes, abrin, ricin, toxins of diphtheria and pseudomonas, bleomycin, gelonin, adriamycin, vinchristine, daunomycin and metothrexate.
10. The cytotoxic drug of Claim 6, wherein A and A' are selected from single aminoacids or polypeptides having from 2 to 20 aminoacids.
11. The cytotoxic drug of Claim 10, wherein said aminoacids are selected from glycine, alanine, serine, leucine, phenylalanine and arginine.
12. The cytotoxic drug of Claim 6, wherein p and o are equal to 1, the backbone polymer being polyasparate.
13. The cytotoxic drug of Claim 6, wherein p=1 and o=2 or p=2 and o=1, the backbone polymer being a copolymer of aspartic and glutamic acids.
14. The cytotoxic drug of claim 6, in which p=o=2, x=0, DM
is OH, DM' is daunomycin or adriamycic and Ay is a Gly-Gly-Leu segment.
15. A method for manufacturing compound I defined in Claim 6, by the reaction of a thiol-copolymer III of formula (III) with a 4-(N-malaiminido)-butyrate derivative II of formula (II) in which formula Z designates the backbone polymer or copolymer indicated in formula I and the symbols EGF, X, m, n and DM are defined as in Claim 6.
16. The use of compounds of formula I as defined in Claim 6 as cyotoxic agents in cancer therapy.
CA000543912A 1986-08-07 1987-08-06 Advanced anticancer therapy and cytotoxic medicaments for its implementation Expired - Fee Related CA1323303C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP86810347 1986-08-07
EP86810347.4 1986-08-07

Publications (1)

Publication Number Publication Date
CA1323303C true CA1323303C (en) 1993-10-19

Family

ID=8196463

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000543912A Expired - Fee Related CA1323303C (en) 1986-08-07 1987-08-06 Advanced anticancer therapy and cytotoxic medicaments for its implementation

Country Status (10)

Country Link
US (1) US5087616A (en)
EP (1) EP0259904B1 (en)
JP (1) JPH01500435A (en)
AU (1) AU608531B2 (en)
CA (1) CA1323303C (en)
DE (1) DE3770730D1 (en)
DK (1) DK184788A (en)
FI (1) FI881579A0 (en)
IN (1) IN165717B (en)
WO (1) WO1988000837A2 (en)

Families Citing this family (289)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IN165717B (en) * 1986-08-07 1989-12-23 Battelle Memorial Institute
US5529932A (en) * 1988-01-28 1996-06-25 Pharmacia, S.P.A. Isolated DNA encoding a plant ribosome inactivating protein from the leaves of saponaria officinalis
IL106992A (en) * 1988-02-11 1994-06-24 Bristol Myers Squibb Co Acylhydrazone derivatives of anthracycline and methods for their preparation
DE4004573A1 (en) * 1989-02-17 1990-08-23 Tanabe Seiyaku Co NEW TRIAL AND HIS PRODUCTION
US5354844A (en) * 1989-03-16 1994-10-11 Boehringer Ingelheim International Gmbh Protein-polycation conjugates
US5576288A (en) * 1989-04-27 1996-11-19 The Salk Institute For Biological Studies Fibroblast growth factor conjugates
US5191067A (en) * 1989-04-27 1993-03-02 The Salk Institute For Biological Studies Fibroblast growth factor conjugates
IL95031A (en) 1989-07-18 2007-03-08 Amgen Inc Method for the production of a human recombinant tumor necrosis factor inhibitor
EP0436005B1 (en) * 1989-07-20 1995-03-29 Sandoz Ltd. Labeled polypeptide derivatives
US5208323A (en) * 1989-08-10 1993-05-04 Universite Laval Coupling of an anti-tumor to an antibody using glutaraldehyde preactivated anti-tumor agent
US6329508B1 (en) 1989-09-07 2001-12-11 Alkermes, Inc. Transferrin receptor reactive chimeric antibodies
US5672683A (en) * 1989-09-07 1997-09-30 Alkermes, Inc. Transferrin neuropharmaceutical agent fusion protein
US5977307A (en) * 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
US5182107A (en) * 1989-09-07 1993-01-26 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5527527A (en) * 1989-09-07 1996-06-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
EP0497867A1 (en) * 1989-10-23 1992-08-12 Schering Corporation Polypeptide inhibitors of gamma interferon
DE69132581T3 (en) * 1990-03-02 2008-07-17 Boston Medical Center Corp., Boston IMPROVED CHIMERIC TOXINS
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
US5137877B1 (en) * 1990-05-14 1996-01-30 Bristol Myers Squibb Co Bifunctional linking compounds conjugates and methods for their production
DE59103985D1 (en) * 1990-05-30 1995-02-02 Deutsches Krebsforsch Polyethersubstituierte tumormittel.
IL98528A0 (en) * 1990-06-21 1992-07-15 Merck & Co Inc Pharmaceutical compositions containing hybrid for killing bladder cancer cells
JPH05508634A (en) * 1990-07-05 1993-12-02 アクゾ・エヌ・ヴエー Receptor-specific toxin conjugate
US5478804A (en) * 1990-09-19 1995-12-26 The Salk Institute For Biological Studies Treatment of tumorigenic pathophysiological conditions with FGF-cytoxic conjugates
WO1992005802A1 (en) * 1990-09-28 1992-04-16 Neorx Corporation Polymeric carriers for release of covalently linked agents
US5431899A (en) * 1991-02-04 1995-07-11 University Of Saskatchewan Rotavirus VP6 as a diagnostic and targeting agent
US5464753A (en) * 1991-03-08 1995-11-07 Univ Illinois Purification and manipulation of bone marrow and blood cells on the basis of P-glycoprotein expression
EP0575545B1 (en) * 1991-03-15 2003-05-21 Amgen Inc. Pegylation of polypeptides
CA2102808A1 (en) * 1991-05-10 1992-11-11 Hanne Bentz Targeted delivery of bone growth factors
EP0529175B1 (en) * 1991-07-22 1995-08-09 BRACCO International B.V. Conjugates chelated with paramagnetic metals for targetting
NZ244306A (en) * 1991-09-30 1995-07-26 Boehringer Ingelheim Int Composition for introducing nucleic acid complexes into eucaryotic cells, complex containing nucleic acid and endosomolytic agent, peptide with endosomolytic domain and nucleic acid binding domain and preparation
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5981273A (en) * 1991-09-30 1999-11-09 Boehringer Ingelheim Int'l. Gmbh Composition comprising an endosomolytic agent for introducing nucleic acid complexes into higher eucaryotic cells
US5922859A (en) * 1992-02-01 1999-07-13 Boehringer Ingelheim International Gmbh Complexes containing nucleic acid which can be taken-up by endocytosis into higher eukaryotic cells
DK0563475T3 (en) * 1992-03-25 2000-09-18 Immunogen Inc Conjugates of Cell Binding Agents and Derivatives of CC-1065
US5679350A (en) * 1992-05-28 1997-10-21 The University Of Toledo Method of delivery of a medicament to a cancer cell using a pathway of plasminogen activator material
EP0643584B1 (en) * 1992-05-28 2000-03-15 The University Of Toledo Delivery of a cytotoxic compound to a cancer cell using a pathway of plasminogen activator material
EP0646174A1 (en) * 1992-06-16 1995-04-05 The Whittier Institute For Diabetes And Endocrinology Recombinant production of saporin-containing proteins
US5393737A (en) * 1992-08-20 1995-02-28 Health Research, Inc. Cytotoxic drug conjugates for treatment of neoplastic diseases
EP0662146B1 (en) * 1992-09-03 2000-07-12 The Regents Of The University Of California Dorsal tissue affecting factor and compositions
US5466672A (en) * 1992-12-04 1995-11-14 Ophidian Pharmaceuticals, Inc. Therapeutic use of clostridium difficile toxin A
US6174530B1 (en) 1993-05-05 2001-01-16 Gryphon Sciences Homogeneous polyoxime compositions and their preparation by parallel assembly
US6001364A (en) 1993-05-05 1999-12-14 Gryphon Sciences Hetero-polyoxime compounds and their preparation by parallel assembly
PT697891E (en) 1993-05-05 2000-09-29 Keith Rose POLIOXIMATE COMPOUNDS AND ITS PREPARATION
US6214345B1 (en) 1993-05-14 2001-04-10 Bristol-Myers Squibb Co. Lysosomal enzyme-cleavable antitumor drug conjugates
US5502037A (en) * 1993-07-09 1996-03-26 Neuromed Technologies, Inc. Pro-cytotoxic drug conjugates for anticancer therapy
US6491905B1 (en) * 1993-09-14 2002-12-10 The Uab Research Foundation Recombinant bacterial cells for delivery of PNP to tumor cells
ES2148259T3 (en) * 1993-09-22 2000-10-16 Hoechst Ag PRO-PROFARMACOS, ITS PRODUCTION AND USE.
US7097839B1 (en) * 1993-10-26 2006-08-29 Thomas Jefferson University ST receptor binding compounds and methods of using the same
US5879656A (en) * 1993-10-26 1999-03-09 Thomas Jefferson University Methods of treating metastatic colorectal cancer with ST receptor binding compounds
US5962220A (en) * 1993-10-26 1999-10-05 Thomas Jefferson University Compositions that specifically bind to colorectal cells and methods of using the same
DE69434279T2 (en) * 1993-12-09 2006-04-06 Centro De Inmunologia Molecular Vaccine containing a human autologous epidermal growth factor and its use
US6551618B2 (en) * 1994-03-15 2003-04-22 University Of Birmingham Compositions and methods for delivery of agents for neuronal regeneration and survival
US5911995A (en) * 1994-08-19 1999-06-15 Regents Of The University Of Minnesota EGF-genistein conjugates for the treatment of cancer
US5587459A (en) * 1994-08-19 1996-12-24 Regents Of The University Of Minnesota Immunoconjugates comprising tyrosine kinase inhibitors
DE4433890C2 (en) * 1994-09-22 1999-02-18 Deutsches Krebsforsch Conjugate of an active ingredient and a native protein that is not considered foreign to the body
DK0788351T3 (en) * 1994-11-10 2003-05-26 Univ Kentucky Res Found Implantable, refillable device for speed-controlled drug delivery directly to an internal part of the body
US5766899A (en) * 1995-02-27 1998-06-16 Board Of Regents , The University Of Texas System Targeted nucleic acid delivery into liver cells
US6015555A (en) * 1995-05-19 2000-01-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US20030119724A1 (en) * 1995-11-22 2003-06-26 Ts`O Paul O.P. Ligands to enhance cellular uptake of biomolecules
ES2195036T3 (en) * 1995-12-22 2003-12-01 Bristol Myers Squibb Co RAMIFIED HYDRAZONE CONNECTORS.
US20050025740A1 (en) * 1996-01-05 2005-02-03 Keith Rose Polyoxime compounds and their preparation
AU735900B2 (en) 1996-03-12 2001-07-19 Pg-Txl Company, L.P. Water soluble paclitaxel prodrugs
US6441025B2 (en) * 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
US6030941A (en) * 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
TW555765B (en) 1996-07-09 2003-10-01 Amgen Inc Low molecular weight soluble tumor necrosis factor type-I and type-II proteins
US5952294A (en) * 1996-07-31 1999-09-14 University Of Pittsburgh Of The Commonwealth System Of Higher Education Peptidyl prodrugs and methods of making and using the same
DE19636889A1 (en) 1996-09-11 1998-03-12 Felix Dr Kratz Antineoplastic transferrin and albumin conjugates of cytostatic compounds from the group of the anthracyclines, alkylating agents, antimetabolites and cisplatin analogues and medicaments containing them
US6080383A (en) * 1997-01-13 2000-06-27 Rose; Samuel Method and composition for the treatment of cancer by the enzymatic conversion of soluble radioactive toxic agents into radioactive toxic precipitates in the cancer
US5965118A (en) * 1997-04-18 1999-10-12 Access Pharmaceuticals, Inc. Polymer-platinum compounds
AU7124598A (en) * 1997-04-18 1998-11-13 Access Pharmaceuticals, Inc. Polymer-platinum compounds
DE69835878T2 (en) * 1997-05-05 2007-04-26 Mayo Foundation For Medical Education And Research, Rochester IGFIIE / IGFBP2 complex
US6916790B2 (en) 1997-05-05 2005-07-12 Mayo Foundation For Medical Education And Research Treatment of osteoporosis
US6548482B1 (en) 1997-05-05 2003-04-15 Mayo Foundation For Medical Education And Research Treatment of osteoporosis
US6030997A (en) * 1998-01-21 2000-02-29 Eilat; Eran Acid labile prodrugs
US6866837B2 (en) 1998-06-05 2005-03-15 Mallinckrodt Inc. Radiolabeled peptides for the diagnosis and treatment of breast and prostate tumors and metastases of such tumors
US6747055B1 (en) 1998-07-17 2004-06-08 The United States Of America As Represented By The Department Of Health And Human Services Water-soluble drugs and methods for their production
WO2000003737A2 (en) * 1998-07-17 2000-01-27 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Water-soluble drugs and methods for their production
GB9904582D0 (en) * 1999-02-26 1999-04-21 Nycomed Imaging As Process
US7018654B2 (en) * 1999-03-05 2006-03-28 New River Pharmaceuticals Inc. Pharmaceutical composition containing an active agent in an amino acid copolymer structure
US6716452B1 (en) * 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US7060708B2 (en) * 1999-03-10 2006-06-13 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US20010041189A1 (en) * 1999-04-13 2001-11-15 Jingya Xu Poly(dipeptide) as a drug carrier
US6706892B1 (en) 1999-09-07 2004-03-16 Conjuchem, Inc. Pulmonary delivery for bioconjugation
US20030054977A1 (en) * 1999-10-12 2003-03-20 Cell Therapeutics, Inc. Manufacture of polyglutamate-therapeutic agent conjugates
US7067111B1 (en) * 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US20020077290A1 (en) * 2000-03-17 2002-06-20 Rama Bhatt Polyglutamic acid-camptothecin conjugates and methods of preparation
EP1274861B1 (en) 2000-03-27 2009-12-23 Thomas Jefferson University Compositions and methods for identifying and targeting cancer cells
CA2410906C (en) 2000-06-02 2012-10-02 Board Of Regents, The University Of Texas System Ethylenedicysteine (ec)-drug conjugates
CA2414650A1 (en) * 2000-06-30 2002-01-10 Board Of Regents, The University Of Texas System Isolation of a cell-specific internalizing peptide that infiltrates tumor tissue for targeted drug delivery
US7163918B2 (en) 2000-08-22 2007-01-16 New River Pharmaceuticals Inc. Iodothyronine compositions
US20020099013A1 (en) * 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
IL155291A0 (en) * 2000-10-16 2003-11-23 Neopharm Inc Liposomal formulation of mitoxantrone
US8394813B2 (en) * 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
WO2002043771A2 (en) 2000-12-01 2002-06-06 Cell Works Inc. Conjugates of glycosylated/galactosylated peptide
CU22999A1 (en) 2001-12-04 2004-10-12 Centro Inmunologia Molecular METHOD OF TREATMENT OF CHRONIC MALIGNAL AND INFECTIOUS DISEASES
EP1389090A2 (en) * 2001-04-26 2004-02-18 Board of Regents, The University of Texas System Diagnostic imaging compositions, their methods of synthesis and use
WO2002092771A2 (en) 2001-05-11 2002-11-21 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
US20100056762A1 (en) 2001-05-11 2010-03-04 Old Lloyd J Specific binding proteins and uses thereof
CA2447335C (en) * 2001-05-15 2011-01-04 Faulk Pharmaceuticals, Inc. Targeted delivery of drugs for the treatment of viral infections
EP1404334A4 (en) * 2001-05-15 2005-02-02 Faulk Pharmaceuticals Inc Targeted delivery of bioaffecting compounds for the treatment of cancer
US7067494B2 (en) * 2001-06-22 2006-06-27 The University Of British Columbia Antimitotic eleuthesides
US7169752B2 (en) * 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US20070066537A1 (en) * 2002-02-22 2007-03-22 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US7375082B2 (en) * 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
US20060014697A1 (en) * 2001-08-22 2006-01-19 Travis Mickle Pharmaceutical compositions for prevention of overdose or abuse
US7338939B2 (en) * 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
US20030109432A1 (en) * 2001-12-10 2003-06-12 Zuo William W. Anticancer polypeptide-metal complexes and compositions, methods of making, and methods of using same
US7261875B2 (en) * 2001-12-21 2007-08-28 Board Of Regents, The University Of Texas System Dendritic poly (amino acid) carriers and methods of use
RU2196604C1 (en) * 2001-12-21 2003-01-20 Северин Евгений Сергеевич Polypeptide displaying analog of receptor-binding epidermal growth factor fragment from 21 to 31 amino acids, its conjugate with doxorubicin and pharmaceutical composition based on thereof
US7700561B2 (en) * 2002-02-22 2010-04-20 Shire Llc Abuse-resistant amphetamine prodrugs
KR20040088519A (en) 2002-02-22 2004-10-16 뉴 리버 파마슈티칼스, 인크. Active Agent Delivery Systems and Methods for Protecting and Administering Active Agents
US7659253B2 (en) * 2002-02-22 2010-02-09 Shire Llc Abuse-resistant amphetamine prodrugs
CN1650024A (en) * 2002-02-22 2005-08-03 新河药品股份有限公司 Use of peptide-drug conjugation to reduce inter-subject variability of drug serum levels
EP2319540A1 (en) * 2002-02-22 2011-05-11 Shire LLC Sustained release pharmaceutical compounds to prevent abuse of controlled substances
US7105486B2 (en) * 2002-02-22 2006-09-12 New River Pharmaceuticals Inc. Abuse-resistant amphetamine compounds
AU2003270778B2 (en) 2002-09-20 2009-10-08 Alpharma Pharmaceuticals, Llc Sequestering subunit and related compositions and methods
CN1723042B (en) * 2002-11-07 2010-12-01 得克萨斯大学体系董事会 Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
EP1627420B1 (en) * 2003-05-07 2020-07-15 Indiana University Research and Technology Corporation Alloyed semiconductor quantum dots and concentration-gradient alloyed quantum dots, series comprising the same and methods related thereto
ES2380622T5 (en) * 2003-05-29 2018-05-30 Shire Llc Abuse resistant amphetamine compounds
US20070087959A1 (en) * 2003-08-19 2007-04-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Method of inducing biomineralization method of inducing bone regeneration and methods related thereof
CA2540678C (en) * 2003-09-30 2011-02-22 New River Pharmaceuticals Inc. Pharmaceutical compositions for prevention of overdose or abuse
AU2004284075A1 (en) 2003-10-22 2005-05-06 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Pyrrolobenzodiazepine derivatives, compositions comprising the same and methods related thereto
EP1694325A4 (en) 2003-12-03 2009-03-25 Univ New Jersey Med Method of preventing survival of retrovirally cells and of inhibiting formation of infectious retroviruses
US9050378B2 (en) * 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
CN100457185C (en) * 2003-12-10 2009-02-04 株式会社东京大学Tlo Coordination complex of diaminocyclohexaneplatinum(II) with block copolymer containing poly(carboxylic acid) segment and antitumor agent comprising the same
ES2338344T3 (en) 2004-04-07 2010-05-06 Rinat Neuroscience Corporation PROCESS OF BONE CANCER TREATMENT THROUGH THE ADMINISTRATION OF AN ANTAGONIST OF THE NEURONAL GROWTH FACTOR.
US20060036084A1 (en) * 2004-08-04 2006-02-16 Lianhua Qu Conglomerated semiconductor nanocrystals
US8454927B2 (en) * 2004-08-04 2013-06-04 Crystalplex Corporation Alloyed semiconductor nanocrystals
CN101203549B (en) * 2005-06-09 2011-04-13 那野伽利阿株式会社 Process for production of polymerized coordination compound of platinum complex
WO2007055902A1 (en) * 2005-11-03 2007-05-18 Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Immunogenic peptides and methods of use for treating and preventing cancer
RU2472812C2 (en) * 2005-12-05 2013-01-20 НИТТО ДЕНКО КОРПОРЭЙШН (Джэпэн/Джэпэн) Polyglutamate-amino acid conjugates and methods
US7820174B2 (en) 2006-02-24 2010-10-26 The United States Of America As Represented By The Department Of Health And Human Services T cell receptors and related materials and methods of use
US9107863B2 (en) 2006-03-27 2015-08-18 The Buck Institute For Age Reasearch Reagents and methods for cancer treatment and prevention
US8758723B2 (en) 2006-04-19 2014-06-24 The Board Of Regents Of The University Of Texas System Compositions and methods for cellular imaging and therapy
HUE032156T2 (en) 2006-06-19 2017-09-28 Alpharma Pharmaceuticals Llc Pharmaceutical compositions
US8088379B2 (en) * 2006-09-26 2012-01-03 The United States Of America As Represented By The Department Of Health And Human Services Modified T cell receptors and related materials and methods
US10925977B2 (en) * 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
MY181496A (en) 2006-10-31 2020-12-23 Government Of The United States Of America Represented By The Sec Dep Of Health And Human Services Smoothened polypeptides and methods of use
EP2102236B1 (en) 2007-01-12 2014-08-06 Government of the United States of America, Represented by the Secretary, Department of Health and Human Services GP100-specific T cell receptors and related materials and methods of use
EP2126127B1 (en) * 2007-01-25 2016-09-28 Dana-Farber Cancer Institute, Inc. Use of anti-egfr antibodies in treatment of egfr mutant mediated disease
US20080181852A1 (en) * 2007-01-29 2008-07-31 Nitto Denko Corporation Multi-functional Drug Carriers
AU2008227123B2 (en) 2007-03-15 2014-03-27 Ludwig Institute For Cancer Research Ltd. Treatment method using EGFR antibodies and src inhibitors and related formulations
CN104800856A (en) * 2007-04-10 2015-07-29 日东电工株式会社 Multi-functional polyglutamate drug carriers
WO2008141107A2 (en) 2007-05-09 2008-11-20 Nitto Denko Corporation Compositions that include a hydrophobic compound and a polyamino acid conjugate
JP2010526917A (en) * 2007-05-09 2010-08-05 日東電工株式会社 Polyglutamate complex and polyglutamate-amino acid complex having plural kinds of drugs
JP2010528122A (en) * 2007-05-09 2010-08-19 日東電工株式会社 Polymer combined with platinum drug
EP2188311B1 (en) 2007-08-14 2016-10-05 Ludwig Institute for Cancer Research Ltd. Monoclonal antibody 175 targeting the egf receptor and derivatives and uses thereof
WO2009029844A1 (en) * 2007-08-31 2009-03-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Compounds for inhibiting wip1, prodrugs and compositions thereof, and related methods
CA2716662A1 (en) * 2008-03-06 2009-03-11 Nitto Denko Corporation Polymer paclitaxel conjugates and methods for treating cancer
WO2010075417A1 (en) 2008-12-23 2010-07-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Survivin specific t cell receptor for treating cancer
WO2010075303A1 (en) 2008-12-23 2010-07-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Splicing factors with a puf protein rna-binding domain and a splicing effector domain and uses of same
WO2010088160A1 (en) 2009-01-28 2010-08-05 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell receptors and related materials and methods of use
WO2010106544A1 (en) * 2009-03-16 2010-09-23 Rajah Vijay Kumar A method and apparatus for - focused resonance nanopermeabilization (forn)
WO2010111617A2 (en) 2009-03-27 2010-09-30 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
US8603814B2 (en) 2009-07-20 2013-12-10 Rutgers The State University Of New Jersey Method of inhibiting nonsense-mediated mRNA decay
US9849146B2 (en) 2009-07-20 2017-12-26 Rutgers, The State University Of New Jersey Inhibition of nonsense mediated mRNA decay by drugs that prevent hypusination of eukaryotic initiation factor 5A
WO2011032099A1 (en) 2009-09-11 2011-03-17 The Board Of Trustees Of The University Of Illinois Methods of treating diastolic dysfunction and related conditions
US20110076232A1 (en) * 2009-09-29 2011-03-31 Ludwig Institute For Cancer Research Specific binding proteins and uses thereof
WO2011041093A1 (en) 2009-10-01 2011-04-07 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-vascular endothelial growth factor receptor-2 chimeric antigen receptors and use of same for the treatment of cancer
WO2011044701A1 (en) 2009-10-16 2011-04-21 The University Of British Columbia Inhibitors of phosphatase and tensin homolog (pten) compositions, uses and methods
WO2011056572A1 (en) 2009-10-27 2011-05-12 The Board Of Trustees Of The University Of Illinois Methods of diagnosing diastolic dysfunction
CA2780572A1 (en) 2009-11-13 2011-05-19 The United States Of America, As Represented By The Secretary, Departmen T Of Health And Human Services Modulated programmed death ligand-1
US8703701B2 (en) 2009-12-18 2014-04-22 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
CA2788304A1 (en) 2010-01-27 2011-08-04 Indiana University Research And Technology Corporation Glucagon antagonist - gip agonist conjugates and compositions for the treatment of metabolic disorders and obesity
DK2547359T3 (en) 2010-03-15 2016-06-06 The Board Of Trustees Of The Univ Of Illionis Inhibitors of beta integrin G-protein alpha subunit-BINDING INTERACTIONS
CN103124740B (en) 2010-09-21 2015-09-02 美国卫生和人力服务部 Anti-SSX-2T cell receptor and associated materials and using method
WO2012054825A1 (en) 2010-10-22 2012-04-26 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-mage-a3 t cell receptors and related materials and methods of use
EP2655401B1 (en) 2010-12-20 2016-03-09 The Regents of the University of Michigan Inhibitors of the epidermal growth factor receptor-heat shock protein 90 binding interaction
EA201390941A1 (en) 2010-12-22 2013-12-30 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн GLUCAGON ANALOGUES, EXPERIENCING ACTIVITY ON GIP RECEPTOR
CN102675422B (en) 2011-03-15 2014-02-05 天津托普泰克生物科技有限公司 Polypeptide drug against hepatitis B virus X protein (HBx)
CN103596981B (en) 2011-04-08 2017-06-16 美国卫生和人力服务部 Anti-epidermal growth factor receptor variant III Chimeric antigen receptors and its purposes for treating cancer
CN102746378B (en) 2011-04-22 2014-10-22 天津托普泰克生物科技有限公司 Anti-fatty acid synthetase polypeptide and its application
BR112013031262A2 (en) 2011-06-09 2016-11-22 Us Health "exotoxins a from pseudomonas with less immunogenic bet cell epitopes, and their use in cancer treatment and prevention, as well as chimeric molecule, nucleic acid, expression vector, cells, pharmaceutical composition and methods to inhibit target cell growth and endotoxin production and pseudomonas and chimeric molecule ".
US20140221283A1 (en) 2011-06-22 2014-08-07 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
EP3392270B1 (en) 2011-09-15 2020-08-26 The United States of America, as Represented by the Secretary Department of Health and Human Services T cell receptors recognizing hla-a1- or hla-cw7-restricted mage
ES2656505T3 (en) 2011-09-16 2018-02-27 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Pseudomonas exotoxin A with less immunogenic B lymphocyte epitopes
RU2644243C2 (en) 2011-10-20 2018-02-08 Дзе Юнайтед Стейтс Оф Америка, Эз Репрезентед Бай Дзе Секретари, Департмент Оф Хелс Энд Хьюман Сёрвисез Chimeric antigenic receptors to cd22
CA2847246A1 (en) 2011-11-17 2013-05-23 Indiana University Research And Technology Corporation Glucagon superfamily peptides exhibiting glucocorticoid receptor activity
AU2013235726B2 (en) 2012-03-23 2017-04-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-mesothelin chimeric antigen receptors
CN104379179A (en) 2012-04-11 2015-02-25 美国卫生和人力服务部 Chimeric antigen receptors targeting b-cell maturation antigen
ES2835200T3 (en) 2012-05-22 2021-06-22 Us Health Medical use of cells comprising anti-NY-ESO-1 T cell receptors
AU2013274078A1 (en) 2012-06-14 2015-01-29 Ambrx, Inc. Anti-PSMA antibodies conjugated to nuclear receptor ligand polypeptides
SG11201408491SA (en) 2012-06-21 2015-01-29 Univ Indiana Res & Tech Corp Analogs of glucagon exhibiting gip receptor activity
ES2602486T3 (en) 2012-06-21 2017-02-21 Indiana University Research And Technology Corporation Glucagon analogs showing GIP receptor activity
US9879065B2 (en) 2012-09-14 2018-01-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services T cell receptors recognizing MHC class II-restricted MAGE-A3
EP2912061B1 (en) 2012-10-24 2019-02-06 The United States of America, as represented by The Secretary, Department of Health and Human Services M971 chimeric antigen receptors
WO2014160627A1 (en) 2013-03-25 2014-10-02 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-cd276 polypeptides, proteins, and chimeric antigen receptors
US9890369B2 (en) 2013-06-20 2018-02-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Cytolethal distending toxin subunit B conjugated or fused to Bacillus anthracis toxin lethal factor
CN105452288B (en) 2013-07-15 2019-08-13 美国卫生和人力服务部 16 E6 T cell receptor of anti-human papilloma virus (anti-HPV)
WO2015051199A2 (en) 2013-10-06 2015-04-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modified pseudomonas exotoxin a
EP3077416B1 (en) 2013-12-06 2019-06-19 The United States of America, as represented by The Secretary, Department of Health and Human Services Thymic stromal lymphopoietin receptor-specific chimeric antigen receptors and methods using same
EP3134514B1 (en) 2014-04-23 2019-09-18 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
EP3149031B1 (en) 2014-05-29 2019-12-18 The United States of America, as represented by The Secretary, Department of Health and Human Services Anti-human papillomavirus 16 e7 t cell receptors
ES2836743T3 (en) 2014-06-02 2021-06-28 Us Health Chimeric antigen receptors that target CD-19
CN107075483A (en) 2014-07-15 2017-08-18 朱诺治疗学股份有限公司 The engineered cell treated for adoptive cellular
TWI805109B (en) 2014-08-28 2023-06-11 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
MX2017005106A (en) 2014-10-20 2017-07-05 Juno Therapeutics Inc Methods and compositions for dosing in adoptive cell therapy.
EP3215164A1 (en) 2014-11-03 2017-09-13 Immures S.r.l. T cell receptors
EP3215601B1 (en) 2014-11-05 2020-05-27 Juno Therapeutics, Inc. Methods for transduction and cell processing
KR20170087514A (en) 2014-12-03 2017-07-28 주노 쎄러퓨티크스 인코퍼레이티드 Methods and compositions for adoptive cell therapy
CA2973964A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
JP6949728B2 (en) 2015-05-29 2021-10-13 ジュノー セラピューティクス インコーポレイテッド Compositions and Methods for Modulating Inhibitory Interactions in Genetically Engineered Cells
MA42895A (en) 2015-07-15 2018-05-23 Juno Therapeutics Inc MODIFIED CELLS FOR ADOPTIVE CELL THERAPY
MA45488A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, KITS AND APPARATUS
MA45489A (en) 2015-10-22 2018-08-29 Juno Therapeutics Gmbh CELL CULTURE PROCESSES, ASSOCIATED KITS AND APPARATUS
RU2021134624A (en) 2015-10-22 2022-03-15 Джуно Терапьютикс Гмбх METHODS, KITS, MEANS AND DEVICES FOR TRANSDUCTION
MA44314A (en) 2015-11-05 2018-09-12 Juno Therapeutics Inc CHEMERICAL RECEPTORS CONTAINING TRAF-INDUCING DOMAINS, AND ASSOCIATED COMPOSITIONS AND METHODS
US11020429B2 (en) 2015-11-05 2021-06-01 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
WO2017096329A1 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
MA43377A (en) 2015-12-04 2021-05-19 Hutchinson Fred Cancer Res METHODS AND COMPOSITIONS RELATED TO TOXICITY ASSOCIATED WITH CELL THERAPY
EP3430549A1 (en) 2016-03-16 2019-01-23 Juno Therapeutics, Inc. Methods for adaptive design of a treatment regimen and related treatments
EP3430548A1 (en) 2016-03-16 2019-01-23 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
AU2017238218A1 (en) 2016-03-22 2018-10-11 Seattle Children's Hospital (dba Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
SG11201809710RA (en) 2016-05-06 2018-11-29 Juno Therapeutics Inc Genetically engineered cells and methods of making the same
EP3463401A4 (en) 2016-06-03 2020-01-22 Memorial Sloan Kettering Cancer Center Adoptive cell therapies as early treatment options
MA45341A (en) 2016-06-06 2019-04-10 Hutchinson Fred Cancer Res METHODS FOR TREATING B-LYMPHOCYTE MALIGNITIES USING ADOPTIVE CELL THERAPY
MA45491A (en) 2016-06-27 2019-05-01 Juno Therapeutics Inc CMH-E RESTRICTED EPITOPES, BINDING MOLECULES AND RELATED METHODS AND USES
CA3028002A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
US20190201414A1 (en) 2016-07-01 2019-07-04 The United States Of America, As Represented By The Secretary, Department Of Health And Human Serv Inhibitor of heme degradation for use to improve antibiotic treatment of mycobacterium tuberculosis infection
WO2018023093A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
MX2019003768A (en) 2016-10-03 2019-06-24 Juno Therapeutics Inc Hpv-specific binding molecules.
EP3548622A1 (en) 2016-12-02 2019-10-09 Juno Therapeutics, Inc. Engineered b cells and related compositions and methods
CN110248678A (en) 2016-12-03 2019-09-17 朱诺治疗学股份有限公司 The method for adjusting CAR-T cell
US20200078400A1 (en) 2016-12-03 2020-03-12 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
JP7429338B2 (en) 2017-01-10 2024-02-08 ジュノー セラピューティクス インコーポレイテッド Epigenetic analysis of cell therapy and related methods
US11517627B2 (en) 2017-01-20 2022-12-06 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
CN110582288A (en) 2017-02-28 2019-12-17 恩多塞特公司 Compositions and methods for CAR T cell therapy
JP7284707B2 (en) 2017-04-07 2023-05-31 ジュノー セラピューティクス インコーポレイテッド Engineered Cells Expressing Prostate-Specific Membrane Antigen (PSMA) or Modified Forms Thereof and Related Methods
AU2018260380A1 (en) 2017-04-27 2019-11-07 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
EP3631468A1 (en) 2017-06-02 2020-04-08 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
JP2020522489A (en) 2017-06-02 2020-07-30 ジュノー セラピューティクス インコーポレイテッド Articles of manufacture and methods for treatment with adoptive cell therapy
CN109517820B (en) 2017-09-20 2021-09-24 北京宇繁生物科技有限公司 gRNA of target HPK1 and HPK1 gene editing method
AU2018345539A1 (en) 2017-10-03 2020-04-16 Editas Medicine, Inc. HPV-specific binding molecules
EP3703688A2 (en) 2017-11-01 2020-09-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
SG11202003657VA (en) 2017-11-01 2020-05-28 Juno Therapeutics Inc Process for producing a t cell composition
SG11202003862PA (en) 2017-11-01 2020-05-28 Editas Medicine Inc Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
EP3704230A1 (en) 2017-11-01 2020-09-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
KR20200116077A (en) 2017-11-01 2020-10-08 주노 쎄러퓨티크스 인코퍼레이티드 Chimeric antigen receptor and coding polynucleotide specific for B cell maturation antigen
CN111556893A (en) 2017-11-06 2020-08-18 爱迪塔斯医药股份有限公司 Methods, compositions, and components for CRISPR-CAS9 editing of CBLB in immunotherapy T cells
MA51210A (en) 2017-12-01 2020-10-07 Juno Therapeutics Inc METHODS FOR DOSING AND MODULATING GENETICALLY MODIFIED CELLS
MA51114A (en) 2017-12-08 2020-10-14 Juno Therapeutics Inc PROCESS FOR PRODUCING A COMPOSITIONS OF MODIFIED T-LYMPHOCYTES
US20210128616A1 (en) 2017-12-08 2021-05-06 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
MA51184A (en) 2017-12-15 2020-10-21 Juno Therapeutics Inc ANTI-CCT5 BINDING MOLECULES AND RELATED METHODS OF USE
CN112292138A (en) 2018-01-22 2021-01-29 西雅图儿童医院(Dba西雅图儿童研究所) Methods of use of CAR T cells
US20210069246A1 (en) 2018-01-31 2021-03-11 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
WO2019170845A1 (en) 2018-03-09 2019-09-12 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
SG11202009284TA (en) 2018-04-05 2020-10-29 Juno Therapeutics Inc T cell receptors and engineered cells expressing same
CA3094468A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
SG11202009446TA (en) 2018-04-05 2020-10-29 Juno Therapeutics Inc T cells expressing a recombinant receptor, related polynucleotides and methods
EP3833759A1 (en) 2018-08-09 2021-06-16 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
JP2021533746A (en) 2018-08-09 2021-12-09 ジュノー セラピューティクス インコーポレイテッド Methods for producing engineered cells and their compositions
JP7394840B2 (en) 2018-08-31 2023-12-08 アンヴェクティ エスアー Chimeric antigen receptor for multiple HLA-G isoforms
US20220002669A1 (en) 2018-10-31 2022-01-06 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
EP3873937A2 (en) 2018-11-01 2021-09-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for g protein-coupled receptor class c group 5 member d (gprc5d)
JP2022512917A (en) 2018-11-01 2022-02-07 ジュノー セラピューティクス インコーポレイテッド Treatment method using a chimeric antigen receptor specific for B cell maturation antigen
EP3877054B1 (en) 2018-11-06 2023-11-01 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
MX2021005734A (en) 2018-11-16 2021-09-10 Juno Therapeutics Inc Methods of dosing engineered t cells for the treatment of b cell malignancies.
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
JP2022513689A (en) 2018-11-30 2022-02-09 ジュノー セラピューティクス インコーポレイテッド Methods for Administration and Treatment of B-Cell Malignancies in Adoptive Cell Therapy
EP3917570A1 (en) 2019-01-29 2021-12-08 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
JP2022526194A (en) 2019-04-10 2022-05-23 エレベートバイオ テクノロジーズ,インコーポレイテッド FLT3-specific chimeric antigen receptor and its usage
JP2022531577A (en) 2019-05-01 2022-07-07 ジュノー セラピューティクス インコーポレイテッド Cells expressing chimeric receptors from the modified CD247 locus, related polynucleotides, and methods.
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
BR112021023411A2 (en) 2019-05-22 2022-02-01 Massachusetts Inst Technology Compositions and methods of circular rna
CN112390894A (en) 2019-08-12 2021-02-23 广东东阳光药业有限公司 Chimeric antigen receptor and uses thereof
JP2023500318A (en) 2019-10-30 2023-01-05 ジュノ セラピューティクス ゲーエムベーハー Cell selection and/or cell stimulation devices and methods of use
CN115398231A (en) 2019-12-06 2022-11-25 朱诺治疗学股份有限公司 Methods related to toxicity and response associated with cell therapy for treatment of B cell malignancies
CN115427550A (en) 2020-01-28 2022-12-02 朱诺治疗学股份有限公司 T cell transduction methods
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
CN115803824A (en) 2020-05-13 2023-03-14 朱诺治疗学股份有限公司 Methods of identifying characteristics associated with clinical response and uses thereof
JP2023527309A (en) 2020-05-19 2023-06-28 オルナ セラピューティクス インコーポレイテッド Circular RNA compositions and methods
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
CN116802203A (en) 2020-11-04 2023-09-22 朱诺治疗学股份有限公司 Cells expressing chimeric receptors from modified constant CD3 immunoglobulin superfamily chain loci, related polynucleotides and methods
KR20230159851A (en) 2021-03-22 2023-11-22 주노 쎄러퓨티크스 인코퍼레이티드 How to Determine the Potency of a Therapeutic Cell Composition
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
CN115404240A (en) 2021-05-28 2022-11-29 上海环码生物医药有限公司 Constructs, methods for making circular RNA and uses thereof
WO2023081735A1 (en) 2021-11-03 2023-05-11 Celgene Corporation Chimeric antigen receptors specific for b-cell maturation antigen for use in treating myeloma
WO2023081900A1 (en) 2021-11-08 2023-05-11 Juno Therapeutics, Inc. Engineered t cells expressing a recombinant t cell receptor (tcr) and related systems and methods
WO2023081526A1 (en) 2021-11-08 2023-05-11 Orna Therapeutics, Inc. Lipid nanoparticle compositions for delivering circular polynucleotides
WO2023164440A1 (en) 2022-02-22 2023-08-31 Juno Therapeutics, Inc. Proteinase 3 (pr3) chimeric autoantibody receptor t cells and related methods and uses
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023230548A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Method for predicting response to a t cell therapy
WO2023231959A2 (en) 2022-05-30 2023-12-07 Shanghai Circode Biomed Co., Ltd Synthetic circular rna compositions and methods of use thereof
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
WO2024031091A2 (en) 2022-08-05 2024-02-08 Juno Therapeutics, Inc. Chimeric antigen receptors specific for gprc5d and bcma

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE38892B1 (en) * 1973-03-28 1978-06-21 Ici Ltd Pharmaceutical compositions
DE3175151D1 (en) * 1980-05-21 1986-09-25 Teijin Ltd Reactive polymer and process for the preparation thereof
IE53166B1 (en) * 1980-08-05 1988-08-03 Searle & Co Synthetic urogastrone gene,corresponding plasmid recombinants,transformed cells,production thereof and urogastrone expression
EP0108132A1 (en) * 1982-05-06 1984-05-16 Applied Molecular Genetics Inc. The manufacture and expression of genes for urogastrone and polypeptide analogs thereof
US4587046A (en) * 1982-05-18 1986-05-06 The Regents Of The University Of California Drug-carrier conjugates
JPS58219124A (en) * 1982-06-15 1983-12-20 Nippon Chem Res Kk Preparation of multiplicative factor of human epithelial cell
US4485093A (en) * 1982-08-13 1984-11-27 Runge Richard G Immunotoxin conjugate which comprises arsanilic acid, useful for treating malignant tumors, particularly pancreatic cancer
JPS59116232A (en) * 1982-12-24 1984-07-05 Teijin Ltd Cell toxicity complex and its preparation
IL71991A (en) * 1983-06-06 1994-05-30 Genentech Inc Preparation of mature human IGF and EGF via prokaryotic recombinant DNA technology
EP0148922A4 (en) * 1983-07-05 1987-06-15 Chiron Corp Hybrid dna synthesis of epidermal growth factor.
JPS6028994A (en) * 1983-07-08 1985-02-14 Wakunaga Seiyaku Kk (21-leucine) human urogastrone, corresponding gene, corresponding recombinant plasmid, transformed cell and their preparation
IL69719A0 (en) * 1983-09-14 1983-12-30 Yeda Res & Dev Synthetic peptides with egf like activity
US4545985A (en) * 1984-01-26 1985-10-08 The United States Of America As Represented By The Secretary, Dept. Of Health And Human Services Pseudomonas exotoxin conjugate immunotoxins
US4522750A (en) * 1984-02-21 1985-06-11 Eli Lilly And Company Cytotoxic compositions of transferrin coupled to vinca alkaloids
CA1283661C (en) * 1984-06-20 1991-04-30 Franz Jansen Imidazolides, process for their preparation and application as intermediates for the synthesis of cytotoxic conjugates
IN165717B (en) * 1986-08-07 1989-12-23 Battelle Memorial Institute

Also Published As

Publication number Publication date
JPH01500435A (en) 1989-02-16
AU608531B2 (en) 1991-04-11
FI881579A (en) 1988-04-06
DE3770730D1 (en) 1991-07-18
DK184788A (en) 1988-06-06
DK184788D0 (en) 1988-04-06
WO1988000837A3 (en) 1988-03-10
US5087616A (en) 1992-02-11
WO1988000837A2 (en) 1988-02-11
AU7788787A (en) 1988-02-24
IN165717B (en) 1989-12-23
FI881579A0 (en) 1988-04-06
EP0259904A1 (en) 1988-03-16
EP0259904B1 (en) 1991-06-12

Similar Documents

Publication Publication Date Title
CA1323303C (en) Advanced anticancer therapy and cytotoxic medicaments for its implementation
Duncan et al. Fate of N-(2-hydroxypropyl) methacrylamide copolymers with pendent galactosamine residues after intravenous administration to rats
Kopeček et al. Water soluble polymers in tumor targeted delivery
EP0701448B1 (en) Amplification of the vitamin b 12 uptake system using polymers
Hoste et al. Polymeric prodrugs
US5869466A (en) Vitamin B12 mediated oral delivery systems for GCSF
US7060498B1 (en) Polycationic water soluble copolymer and method for transferring polyanionic macromolecules across biological barriers
Nori et al. Intracellular targeting of polymer-bound drugs for cancer chemotherapy
EP0187547B1 (en) Synthetic polymeric drugs
AU691225B2 (en) Improved interferon polymer conjugates
JP2002543111A (en) Amplification of folate-mediated targeting to tumor cells using polymers
JP2003506319A (en) Vitamin-related dual targeting therapy
Nori et al. Subcellular trafficking of HPMA copolymer–Tat conjugates in human ovarian carcinoma cells
Van Heeswijk et al. The synthesis and characterization of polypeptide-adriamycin conjugates and its complexes with adriamycin. Part I
JP4718117B2 (en) PH-sensitive polymer conjugates of anthracycline anticancer drugs for targeted therapy
US20050220754A1 (en) Vitamin directed targeting therapy
MXPA01013276A (en) Biologically active materials.
JPS62167800A (en) Ribosome deactivated glycoprotein modified by oxidation of oxide unit and formation of schiff base and in vivo lasting immunotoxin
EP0485749A2 (en) Chemical modification of antibodies for creating of immunoconjugates
Franchini et al. Perspectives on: Recent advances in poly (amidoamine) s chemistry
Hoes et al. Design of soluble conjugates of biodegradable polymeric carriers and adriamycin
SK278506B6 (en) THE POLYMERIC DRUG AND PREPARATION METHOD THEREOF m cells.
Lloyd et al. Targeting and lysosomal handling of polymethacrylamide-oligopeptide conjugates
Fenili NOVEL BIOACTIVE AND BIODEGRADABLE BIOMATERIALS OF POLY (AMIDOMINE) STRUCTURE
AU2004201277A1 (en) Amplification of folate-mediated targeting to tumor cells using polymers

Legal Events

Date Code Title Description
MKLA Lapsed