CA1330295C - Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas - Google Patents

Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas

Info

Publication number
CA1330295C
CA1330295C CA000578596A CA578596A CA1330295C CA 1330295 C CA1330295 C CA 1330295C CA 000578596 A CA000578596 A CA 000578596A CA 578596 A CA578596 A CA 578596A CA 1330295 C CA1330295 C CA 1330295C
Authority
CA
Canada
Prior art keywords
drug
effective amount
therapeutically effective
pharmaceutical composition
agent
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
CA000578596A
Other languages
French (fr)
Inventor
Edward E. Luck
Dennis M. Brown
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.)
Intarcia Therapeutics Inc
Original Assignee
Matrix Pharmaceutical 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 Matrix Pharmaceutical Inc filed Critical Matrix Pharmaceutical Inc
Application granted granted Critical
Publication of CA1330295C publication Critical patent/CA1330295C/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
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

TREATMENTS EMPLOYING VASOCONSTRICTIVE
SUBSTANCES IN COMBINATION WITH CYTOTOXIC
AGENTS FOR INTRODUCTION INTO CELLULAR LESION AREAS

ABSTRACT OF THE DISCLOSURE
A pharmaceutical composition and method of treating cellular disorders involving abnormal solid cellular growths which comprises administering a pharmaceutical composition containing cytotoxic agents in combination with a vasoconstrictive drug. Enhanced effectiveness of the composition is observed, with reduced cytotoxic effects on cells distant from the site of introduction. Agents may be included to enhance therapeutic gain and reduce adverse affects to normal tissue.

Description

1 330295 24619/;~A~1-2 ~ ~
BIR07:09/23/87 ~ ~-~.:
tBEATMENTS EMPLOYING VASOCONSTRICTlVE
SUBSTANCES IN COMBINATION WIT~ CYTOTOXIC
AGENTS FOR INTRODUCTION INTO CELL~LAR LESION AREAS

The treatment o~ many cellular di~orders, for ~-;
example, tumors, and hyperproll~erative diseace~ e.g.
psoriasis, involves the use o~ cytotoxic drugs. These drugs exert their activity in a variety of ways, u~ually interfering with a cellular ~unction essential ~or the repl$cation and/or viability of the cell. In ~; many, if not most inqtances, the drug i~ not ~pecLflc for the abnormal cell, but rather tends to exert its effectivene~s due to the more rapid proli~eration of the abnormal cell, a~ compared to normal cells. Whlle many organ~ o~ the body of a mammalian ho~t regenerate : cells rather ~lowly. there are alqo other organ~, par-ticularly bone marrow, which ln~ol~e rapld prolifera-; 25 tion o~ Jtem cell~. Therefore, the cytotoxic agents not only can detrl~entally affect the Jlowly regen-;~ erating cella, but al~o ha~e a particularly pernicious ffect on the i~cuno ~yat-m.
Deaplte the many di~ad~antage~ and ~ide 3 ~ects o~ employlng the ~trongly cytotoxic drugs, they have ~ound extenal~e appllcatlon becau~e they have pro-~ided poqitl~e re~ult~. Howe~er, there i~ ~ub~tantial intere~t ln belng able to employ the drug~ in a manner which dir~ct~ thelr activity toward the abnormal cellq, whlle simultaneously protecting ~en~$Si~e normal cells, both iQ the ~lcinity o~ and distant from the ~ite of application. ;`

. ~ .

, ~

- ::
1 3302q5 Relevant Literature U.S. Patent Nos. 4,322,398. 4,347,234, 4,349,530, 4,391,797 and 4,536,387 describe implants and controlled release of drugs. Implantation of drugs in lesions is described in Maugh, Science (1981) 212:1128-1129: Macek et al.. Abstracts of Immunoloay. 4109, p. 1053: Miyata et al. Cancer Research (1983) 43:4670-4675; McLaughlin et -al. Cancer Research (1978) 38:1311-1316: and Bier et al.
Cancer (1979) 44:1194-1200. U.S. Patent No. 4,619,913 describes the use of cytotoxic drugs and vasoconstrictive drugs in a proteinaceous matrix. See also references cited therein.

Abnormal solid cellular growth, particularly tumors or adjacent tissue that may contain tumor cells, and other hyperproliferative conditions, are treated by subjecting the abnormal growth area or tissue suspected of containing tumor cells to a pharmaceutically effective amount of a combination of a cytotoxic drug and a vasoconstrictive drug. The combination substantially inhibits migration of the cytotoxic drug from the site of application by alteration of the blood flow serving the tumor/lesion area, so as to maintain the primary effect of the cytotoxic drug at the site of application.

Accordingly, and in a first aspect, the present invention i8 a pharmaceutical composition consisting essentially of a therapeutiaally effective amount of a vasoconstrictive drug, a therapeutically effective amount of a cytotoxic drug and a protein-free, aqueous, ~-~
pharmacologically acceptable vehicle.
:.
In a further aspect, the invention is the use of a ;~
composition consisting essentially of a uniformly dispersed vasoconstrictive drug, present in a therapeutically effective amount, and at least one uniformly dispersed cytotoxic drug present in a therapeutically effective amount, in a non-proteinaceous ;, -~, L

; ` 1 330295 2a aqueous medium to treat a neoplastic lesion or surrounding tissue, whereby said drug is retained in the ~ :
immediate environment avoiding significant levels of the drug at sites distant from the site of application.

The treatment may be employed with various solid tumors, including carcinomas, lymphomas and sarcomas.
The chemotherapeutic compositions comprise an antiproliferative or cytotoxic drug and a -' '~

~ .
, .. ..
,'~
, ,~:

,: L,i: ~;

vasoconstrictive drug. The drug combination is applied ~-~
at the region of the lesion either intralesionally by injection or adjacent to or at the surface of the leæion, conveniently by means of a catheter.

It will be understood that the term cytotoxic refers to any anti-neoplastic or anti-proliferative agent. It will also be understood that the term vasoconstrictive --refers to any agent which constricts capillaries 80 as to substantially restrict the flow of blood to or through the blood vessel. ~y employing the subject combination, the circulating blood level of the cytotoxic drug remains low. In this way, an enhanced therapeutic gain is achieved, that is, the cytotoxic effect on the treated neoplastic cells is greater as compared to susceptible normal cells remote from the application site.

Hyperproliferative growth means a preneoplastic lesion or condition such a~ psoriasis, keloids, or warts.
It will be understood that the terms neoplastic and neoplastic lesion refer to any new or abnormal growth, such as: cancer, oncogenically transformed cells, carcinomas, melanomas, lymphomas, myelomas, both benign and malignant tumors, and sarcomas.
~: ', ' ,.
Illustrative of the various diseased states or therapeutic modes in which the subject invention may find application are: ~1) Neoplasms in which local recurrence is typical and drug bioavailability is compromised. e.g., brain: (2) tumors in which suspected neoplastic cells remain in the tumor bed following surgical resection, e.g., breast: (3) tumors which are poor candidates for surgical or radiation management, e.g., head, neck, prostate, etc.: (4) adjunctive tumor therapy in combination with physical or non-chemical treatments, e.g., radiation and/or hyperthermia: (5) ; 35 hyperproliferative diseases refractory to conventional I therapy, e.g., psoriasis: (6) concurrent treatment with !~: B

4 ~ 330295 syctemic chemotAerapy; an~ (7) concurrent with systemlc rescue, e.g. intra~enoua leucororin.
In Come inqtance-q it may be ~e~irable to Include non-protelnaceou~ carrier~, qo a~ to Form a 5 liquid, particularly aqueou~, or semi-~olid or gel -~
medium. Compositions which ~ay ~ind use are ph~io-logically acceptable substances, ~uch as carbohydrates, polylactate, agaroses, dextrans, c_lluloce, gums, etc. Synthetic peptides ~ay ~ind uqe, such as polylysine, polyarginine, etc~ The composition ~ay be formulated with lipid~ to form liposomes or in a solid ~orm in combination with ~ilicones, epoxide resinC, hydroxyapatite, etc. The drugs and carrier will be selected to minimi~e any inacti~ating effects on the drugS-Various drugs may be employed which are usedin chemotherapy and act a~ alkylating agentq, enzyme inhibitors, prolif~ration inhibitors, lytic agents, DNA
~ynthe~i~ inhibitors, membrane permeability modifiers, DNA intercalators, antimetabolites, or the like.
Illu~tratlre drug~ include: ci~platin (Platinol)*, doxorubLcin hydrochloride (Adriamycin), bleomycin - qulfate (31enoxane), fluorouracll, ~incrl~tlne sul~ate ;~
(Oncovin), ~lnbla~tine ~ulfate (Velban) VP-t6, -~
25 chlorambucil ~Leukeran), melphalan ~Alkeran), buqul~an ~`~
tMyleran), carmu~tine ~8CNU] (BlCNU), lomu~tlne ~CCNU~
tCeeNU), ~treptozotocin, thiotepa, da¢arbazlne ~DTIC-DOME), methotrexate, ¢ytarablne (Cytoaar-U), azarlblne, mercaptopurine ~Purinethol), thio6uanine, actino-3 mycin D, plicamycin (Mithracln), ritomycin-C (Muta-mycin), asparagina~e MSD ~El~par)~ procarbazine hydro-chloride ~Matulane), predni~one, prednl~llone, triam-clnolone, te~to~terone, e~trogen, in~ulin~, and hydroxyurea (Hydrea). Other druga of lntere t include radio~ensitizer~, ~uch a~ SR-2508 and mi~onLdazole;
hyperthermia sen~itlzer~, ~uch a~ lidocalne and marcaine; bloreductl~e agent~, ~uch aa mltomycin-C
: * Trade Marks are in brackets.

~ . ' .. ', . , . ' ~ ;;;

~"` 5 1 33 02q 5 benzotriazine dioxi~e-e and nitroheteroCyCllC compound3 quch aa benzni~azole. See Carter and Li~lng-~ton, Drug Ava~lable to Treat Canc~r, In Principlee of Cancer Treatment (Carter _ al., ed-q.) Chaptee 10, pp. 11 t 145, 1982, McGra~-Hill, Inc., N.Y.
The druss may be used individually or in com-bination, depending upon the nature oF the drug, the ;~
tumor, and whether cooperative action i5 pharmacolog-ically indicated. The drus composition can be ~urt~er 10 modi~ied, by modi~y~ng the drug, particula~ly by bonds ~-which allow for enzymatic clea~age, e.g., hydrolysis, or by lntroducing materials into the composition which will further aid in the maintenance of the retention o~
the drug at the site o~ introduction.
Illustrative vasoconstrictive agents are~
(1) ~ympathomimetics including the catecholamlnes, norepinephrlne, epinephrine, l~oproterenol, dopamine, and related compoundq ~uch as ephedrine and other phenylisopropylaminea, phenylephrine, amphetamine, metraminol, methoxamine; (2) ergot alkaloid-q including lyaergic acid, lyaergic acid diethylamine, ergonovine, m(ethylergonavine, methy~ergide, ergotamine; (3) the angiotenaina; and (4) the proqtaglandina. Vaaocon-atricti~e agenta are descrlbed in Medical Pharmacology ;
~1984), C.V. Moaby, Company, Chapter 15.
For enhancing cytotoxicit~, other agents -~uch aa thoae af~ecSing tiaaue architecture may be uaed.
~hoae lncludo onzymea which can InJur~ the atroma, auch aa the poptida~e~ papain, chymopapaSn, trypaln, a~ylase, coliagenaae and chymotryp~ln. Slmilarly, agenta af~ecting cellular permeabllity may be employed, auch aa anionlc, cationlc or non-ionlc detergenta, a.g., Tween 80, amphotericin B, dlmethylsulfoxide.
~naeathetica, ~uch as procaine, may be included in the 35 formulations. Al~o, other agent3 ~hich o~ercome ;~
inherent cellular drug rea~atance, like verapamil, may ~ be included.
;:~

~ llr~l-. ~ - . ~- ,.... ;

In addition, the drug(~) can be employed encapsulated in liposome~ or other controlled rate release composition~ ~o a~ to provide ~or separate and dlstinct rates o~ release of the drug. In this way, 5 multiphasic compositions can be prepared, so as to provide ~or sustained release 3~ the drug over long periods of time. Formation o~ liposomes with inclusion of various materials is described in Papahadjopoulos (1978) Annal3 o~ t~e N.Y. Academy o~ Science, 308;
Gregoriadis and Allison tl980) Liposomes in 3iological Sy~tem , John Wiley and Sons, Leserman et al., Nature (1981) 293:226-228; Barhet et al., Supramol. Struct.
Cell. Bio. Chem. ( 1981) 16: 243-258; and Heath et al., Science S1980) 255:8015-8018. Alternatively, other 5 method~ o~ encapsulation can be employed where the drug is encapsulated in a biodegradable substance, where the rate o~ release ia related to the thicknecs of the -biodegradable coat.
For enhancing cytotoxic act~vity, various ad~uvant materials may be incorporated into the phar-maceutical composition, such a~ radioactive pellet3, s.g., radionuclides technicium or iridium; radiation sensitizers, e.g., nitroimidazoles and halogenated pyrimidlnes (BUdR); repair inhibitors, e.g., methylated xanthines; bioreductive agents, which are actiYated onLy in hypoxic cells; cytoklnes, such as the - lnter~erons, lymphokinea, ~uch as interleuk$n-2; tumor growth lnhlbitors, ~uch as tumor necros$a factor, tran~form$ng growth ~actor-B, etc. Angiograph$c 3 contrast media may also be incorporated into the compos$t$on to ~ac$1itate viaualization.
Other mater$als aa well may be included to enhance an $mmunogen$c cytotox$c response, e.s., pro^
liferat$on and invasion of macrophage, helper T-cells, ~ 35 etc. Illu-~trati~e adjuvant~ include Corynebacterium i; parvum, aacillus Calmettc-Guerin cell wall or cell wall :; akeleton preparationa, Mycobacterium bovi_ strain, ~ 7 1 3302~5 etc. See ~lyata et al., Cancer Res. (1983) 43:4570-4675; Bler et aI. Arch. Otorhinolaryngol. (1982) 236:245-255; and Mehani~hlin et al., Cancer Res. t1978) 38:1311-1316, The pharmaceutical compo~ition o~ this inven-tion may be un$~ormly dispersed in a physiologically ;
acceptable medium, particularly aqueous, 3uch as qaline, phosphate ~uf~ered saline, distilled water, etc. The aqueous medium will be ~uf~icient to providefor an amorphous di~persion, usually a 301ution, capable of flowing under mild pres~ure. Usually, the -~
liquid aqueou3 medium will be at least 5 weight percent of the entire composition, more usually at least 10 -~
weight percent, and not more than about 75 ueight percent, usually not more than about 50 weight percent, ~o as to provide a ~lowable mixture. The amount will ~ary depen~ing upon the nature of the drug(s), the nature of any carrier materlal, the presence of other ~aterial~, and the like. The concentration of protein other than carrier protein in the aqueous medium, if ~ `~
any, will range from about O to 75 mg~ml.
The cytotoxlc drug(s) vlll normally be a liquid or ~olld, or pro~lded in a flowable di~peraed 25 ror~ and ~Ill generally range from at lea~t about 0.1S
by weight up to about 50: by uelght, more u~ually beInt from about 1S to 50~ by welght Or the rormulatlon. The ~a~ocontrlcti~e agent(~) ~111 genorally be pre~ent In about 0.5 to 35, u-~ually 5 to 20 weight percent of the formulation.
Other anclllary addlti~e~ or agent~ a~
de~cribed abo~e may be pre~ent and ~ill rary in total amount from about 0.005 to 15 ~eight percent, u~ually from about 0.01 to 10 welght percent Or the total compo~ition.
In addltion to the cytotoxlc drug~ and ~aso-con~trlctor~, a number of minor components may al~o be a 1 330295 included ~or a va~i~tl o~ pur~oses. Tnese agents will ~or the most part impart properties which protect the stability of the composit~on, control the pH, or t~e like. Illustrative agents include pho~phat~ or acetate buf~ers, methyl or propyl paraben, polyethylene ~ly-cols, etc. These a3ent3 3enerally will be present in less than about 2 weisht percent of the total composi-tion, u_ually less than about l wei~ht percent, an~
individually may vary ~rom about 0.001 weight pe~cent 10 to about 1 weight percent. ~:
Aq alr_ady indicated, in ~ome instances the drug will be encapsulated particularly in liposomes.
Liposomes are prepared from a variety o~ lamellar-forming lipids including phospholipids, e.g., phospha-tidylcholine, phosphatidylethanolamine, etc., ganglio-aides, sphingomyelins, steroidq, e.g., cholest~rol, etc. Usually, the weight of the lipids in relation to the weight of drug will range ~rom 1 to 5 l1ter~ of entrapped dru~ per mole of amphipathic liPid.
The pharmaceutical composition of the preaent invention can be prepared by combining the variou_ com-ponent3 in a sterile enYironment. The composltion wlll be provided in a convenient form, usually admixed with at least a portion of the total aqueou-~ medlum to be employed. The composition will be quf~iciently work-able 90 that, upon admixture of the other agentq, a unlform diqperaion can be obtalned.
Other materlal~, as appropriate, may be added concomltantly or ~equentially. After ensuring the uni-3 ~orm disper~ion of the various components in the mix-ture, the mixture may be sterilized and 3ealed in an appropriate container. In the erent the various com-ponents are unstable or form undesirable Complexes when stored in a mixture prior to admini~tration, each com-ponent may be diapen~ed at an appropriate concentration ~ into a 3eparate container for mixlng ~ust prior to -~ -~
i.~ adminiqtration. Those components which are ~table -~

~ , -.' :- ~,' ` 9 1 330295 together may be dispensed togethe~ into a ~ingle con~
tainer ~or mixture with one o~ more reagentq containin those additional insredients ~ound to promote instabil- ~-ity or to ~orm undesi.able co~plexes. A device or kit containing ~eparate components may be prepared which facilitates easy Formulation prior to adminiatration.
The concentration oF each separate component is farmu-lated so that the therapeutically e~ective concentra-tion o~ each agent is achieved when all the separate components in the kit are admixed.
The compoqition o~ this in~ention can be used in the treatment Oe a wide variety of neopla~tic or benign le~ions. Illustrative tumors include carcinomas, sarcomas and melanomas, such as ba~al cell carcinoma, squamous cell carcinoma, melanomas, soft tisque sarcoma, ~olar keratoses, Kaposi'3 3arcoma, cutaneou_ malignant lymphoma, Bowen's di3ease, ~ilm'~
tumor, hepatomas, colorectal cancer, brain tumor~, myco-qi3 fungoides, Hodgkin~ lymphoma, polycythemia vera, lymphomas, oat cell sarcoma, etc. Al30 sub~ect to treatment are wart~, ~enign lesions, preneopla3tic lesions, and hyperproliferatire diseases, such a~
psoriasis.
The composition of thi3 invention will be administered to a tumor to provide a cytotoxic amount o~ drug at the tumor ~ite. The amount of cytotoxlc drug admlnistergd to the tumor ~ite wlll generalIy range from about 0.1 to 50~, more usually about 0.5 to 300 mg/kg o~ ho~t, depending upon the nature of the 3 drug, size of tumor, and other conslderations.
In view of the wide diversity of tumors, nature of tumors, effective concentration~ o~ drug, relati~e mobility and the like, a definitive amount for ~ administration cannot be specified. With each drug in ; 35 each tumor, experience will provide an optimum level.
One or more administration~ ~ay be employed, depending upon the lifetime of the drus at the tumor site and the - ' 1 330295 response o~ the tumor to the drug. Administ.~tion may be by ~yrin~e, cathetsr or other convenient means a1lowing for application ~f a flowable compo~ition at the tumor sita. Administration may be every third day, wee~ly, or les~ ~requent, such as ~iweekly or at monthly intervals.
Injection may be at one or more ~ites depending on the size o~ the lesion. Nesdles of about 1-2 mm diamete~ are con~enient. For multiole injec-tion, templatPs ~ith predrilled holes ~ay be employed.
The method o~ the present invention finds par-ticular advantage with tumors or lesions which are clinicalIy relevant. The compositions provided show particular therapeutic gain with tumors greater than 100 mm3, more particularly, greater than 150 mm3, in volume, a~ well as flat cutaneous lesion~ with surface areas ranging from 10 mm2 to 100 cm2 or greater.
The method of the present invention also reduces local inflammation as a result o~ drug admini-gtration, therefore, local or adjacent tiqsue i9 le~slikely to be affected by the drug. Furthermore, due to the low migratory level of the drug from the 3ite of ;placement, higher drug do~age~ can be admini~tered to the alte without adverse affects to normal tiqsue diqtant from the placement _ite or to lymphocyte_.
The method Or the preaent lnvention findq ,ad~anta~e ln conJunction with other ~orm~ of therapy.
he leqlon~ may be irradiated prior and~or ~ubqequent `
to drug admlniqtration. Dose rates may vary from about 20 to 250 rad/min, usually 50 to 150 rad/min, depending on the le~ion, period of expo~ure, and the like.
Hyperthermia (heat) may be uqed as an adjunctive treat-ment. Treat3ent ~ill u~ually involve heating up of the tissue to be treated to about and including 43~C for about 5 to 100 min.
In order to demon~trate the _ub~ect invention the following inveqtigationa were performed. The `

,1 1 330295 ~ollowing experiments are illust.~tive of ~ariol~s emb~diments of the invention, but do not in any way limit it.
The transplantable expecimental murine ~ibro-sarcomas RIF-1 or KHT were grown intradermally (2 x 105 cells injected i.d.) in the ~lank of 5 ~onth old male C3H mice. When tumors reached a volume o~ -100 mm3 t~ey were used ~or the studias. The cytotoxic drugs under study (all were ~ormulated ~or human clinical use) were made up just prior to uqe in thei~ appro-priate aqueouq vehicle except those experimental groups in which epinephrine-HCl (1 mg/ml) was added to the aqueous drug (usually _10d o~ total volume). Drug concentrations were: adriamycin, 3.75 mg/ml; BCNU, 10 mg/ml; vinblastine, 1.2 mg/ml; and 5-fluorouracil, 30 mg/ml. The drugs w~re adminiqtered intra-pe~itoneally (i.p.) or intratumorally (L.t.) in an in~ection volume of 0.1 ml. The tumorq of the treated mice as well a~ untreated control tumor3 were measured three times per week by vernier calipers and their ~olumeq were calculated by the formula:
.
: V J ~/6 x D1 x D2 x D3 .'~ ' where D1-D3 are tumor diametar~ in mm. The tlm~ (days) for the tumors to 3row to four timea their origi~al ex-; perimental volume wa~ u~ed a~ a parameter Oe treatment ~; erfecti~eness. Epinephrlne in the doses u~ed (1 mg/kg) wa~ not chemotherapeutlc ~hen u~ed alone. The data for 3 the drugs studied i9 summarized in Table 1.
Further evldence for the demonstration of the ~nhancement of localized tumor concentration of chemo-!` ~ therapy drugs in combination ~ith a vasoconstrlctive agent (such a~ eplnephrine) compared to the tumor concentratlon of the chemotherapeutic agent used alone i9 demonstrated in Table 2. In thi~ experiment, mice ~-- bearing the RIF-1 fibrosarcoma~ are in~ected ~ ~ .

' r ~

' ~

12 1 330295 ~ ~

Table 1 ~n~luence of Epinephrine (1 mg/k~) (Epi) In C~mbination with Anticancer Drugs Administ_red Intratumorally (i.t.) on the Growth o~ Murine Fibrosarc~mas .
Dose Route o~ Tumor Gr~w~.
Drug(mg/kg 9W) Administr~tion T~m~r Ratio*
Adriamycin 15 i.p. RIF-1 1.63 Adriamycin 15 i.t. RIF-1 3.04 Adriamycin 15 i.t. RIF-1 3.24 + Epi _ 5-Fluorouracil 100 i.p. RIF-1 1.20 5-Fluorouracil 100 i.t. RIF-1 1.30 5-Fluorouracil 100 i.t. RIF-1 1.44 Epi ~ ~ .
~- 20 Vinblas~ine 2 i.P- KXT 1.16 Vinblastine 2 i.t. KHT 1.76 -~inbla~tine 2 i.t. KHT 2.92 Epi Vinbla~tine 2 l.p. RIF^1 1.12 Vinblastine 2 i.t. RIF-1 3.01 ; 25 Vin~lastine Z i.t. RIF-1 4.00 Epi ~` 3CNU (carmu~tlne) 40 i.t. KHT 2.08 I; BCNU ~ Epi 40 i.t. KHT 2.~0 * Thi~ ratio indicated the time it would tak~ a treated tumor to ~; grow to 4x its initial ~lze, d~rided by the time it would take an untreated tumor to grow to 4x its initial ~ize. For example, if i~ took a treated tumor 40 day~ to grow ~rom 100 ~ mm~ to 400 mm~, a4d an untrsated tumor 10 days to grow from ':~ 100 mm3 to 400 m~; thi~ would glve u~ a tumor growth ratlo of 4Ø Thw , increasing ratio~ indlcate improved treatment e~ectivene~s.
,,~"~

.

~ ~ " f ' 1 3302q5 Table 2 Influence o~ Epinephrine (Epi) (0.5 mg/Xg) on Intratumoral Deli~ery of 5-FU: Ratios o~ Tumor to Plasma Concentration and Ratios of 5-FU Concentration ~ Epi in RIF-1 Tumor ... ~

5-FU Tumor/Plaqma Ratio Time Post Injection 5-FU, 5-FU ~ Epi, Tumor (5-FU ~ Epi, i.t.) (Hours) i.t. i.t. Tumor (5-FU, i.t.) 0.5 24.59 198.34 4-94 1,0 14.63 137.42 5.04 2.0 47.53 82.35 1.68 4.a 19.01 58.46 2.24 .
24.0 4.10 6.57 1.48 - ~-. 25 ::
.: :
., , :. .
'`::

~ 35 ;. ~
'',' . ~.

~: .

~ ' 9 i . 14 l 330295 intratumoeally (i.t.) with 0.1 ml o~ an aqu~ous -~olution of tritium labele~ 5-fluorouracil (5-FU) (25 ~Ci/~l qtoc~ ~olution, Amer~ham), or an equivalently radioactively labeled _olut~on of 5-~U
that alqo containq epinephrine at a ~inal concentration of 2.5 mg/ml. Plaqma and t~mor 3amples ~ere taken 0.5, 1, 2, 4 and 24 hours aFter in~ection. The tiqsueq were ~olubilized in Scintiges~M(Fis.~er Scientific), corrected for their tissue wei~ht, and 200 ~l o~ the solubilized samples were added to 6 ml of ~cintillation fluid and counted in a liquid scintillation counter for the pre~ence o~ tritium. The values in Table 2 are ratios of the normalized counts (based on 100 mg of tissue, wet weight) in tumor divided by the radio- ~ ~
15 activity in the plasma samples. The data demonstrate -that at erery time point evaluated, a 1.48 to 5.04 -~
enhancement of 5-FU tumor concentration to plasma -concentration ~as obserxed when epinephrine wa~
included in the 5-FU. Thiq lndicate~ that the ra~oconstricti~e actir~tg of epinephrine promote~ the localization and retention of the drug in the tumor and llmit-q the dlffuaion Or the cytotoxic agent from the -tumor into ~yatemic circulation (i.e. lov plaqma drug concentrationa).
Thu~, it can be ~een that improred neopla~tlc therapy l~ achlered by applying to a neopla~tlc le~lon a compo-~ition comprlaing a cytotoxic drug In combina-tlon wlth a ~a~ocon~trlctire a8ent. It i~ found thtt by employlng the drug compositlon, greatly enhanced 3 localized drug concentration can be achiered. In addition, in rlew of the _igniflcant cytotoxic ef~ects of drugi~ employed in chemotherapy, _y~temlc expoaure is aub~tantially timinl hed. Therefore, higher lerels of cytotoxlc drug~ can be employed at the ~ite of inter-35 e~t, wh$1e the remainder of the ho~t i~ not expo~ed to ~-ignlficant lerel~ of the drug.

:
~ 1 330295 Althou3h the foregoing invention has been described in some detail by way of illuatration and :
example ~or purposea o~ clarity o~ understanding, it will be obvloua that certain changes and modificationq may be practiced within the scope of the appended claims.

', : ' :: -. ~5 ,..

. 3 ~:

~; 35 ~, .,,~ .

I :~
:` :
1~`

Claims (20)

1. A pharmaceutical composition consisting essentially of a therapeutically effective amount of a vasoconstrictive drug, a therapeutically effective amount of a cytotoxic drug and a protein-free, aqueous, pharmacologically acceptable vehicle.
2. A pharmaceutical composition according to claim 1, wherein said vasoconstrictive drug is at least one of the following: epinephrine, norepinephrine, the catecholamines, epinephrine, isoproterenol, dopamine, ephedrine and other phenylisopropylamines, phenylephrine, amphetamine, metraminol, methoxamine, ergot alkaloids, ergonovine, methylergonavine, methysergide, ergotamine, the angiotensins and the prostaglandins.
3. A pharmaceutical composition according to claim 1, wherein said cytotoxic drug is at least one of the following: an alkylating agent, enzyme inhibitor, lytic agent, DNA synthesis inhibitor, DNA intercalator, or antimetabolite.
4. A pharmaceutical composition according to claim 1, wherein said cytotoxic drug is at least one of the following: cisplatin, doxorubicin hydrochloride, bleomycin sulfate, fluorouracil, vincristine sulfate, vinblastine sulfate, chlorambucil, melphalan, busulfan carmustine, lomustine, streptozotocin, thiotepa, dacarbazine, methotrexate, cytarabine, azaribine, mercaptopurine, thioguanine, actinomycin D, plicamycin, mitomycin-C, asparaginase, procarbazine hydrochloride, prednisone, prednisilone, triamcinolone, testosterone, estrogen, insulin, BUdR, nitrosourea, carmustine, lomustine, and hydroxyurea.
5. A pharmaceutical composition according to claim 1, in addition consisting of an agent affecting tissue architecture and present in a therapeutically effective amount.
6. A pharmaceutical composition according to claim 5, wherein said agent affecting tissue architecture is at least one of papain, chymopapain, trypsin, amylase, collagenase and chymotrypsin.
7. A pharmaceutical composition according to claim 1, in addition consisting of an agent affecting cellular permeability and present in a therapeutically effective amount.
8. A pharmaceutical composition according to claim 7, wherein said agent affecting cellular permeability is at least one of detergents amphotericin B and dimethylsulfoxide.
9. A pharmaceutical composition according to claim 1, in addition consisting of an agent affecting said cytotoxic activity and present in a therapeutically effective amount.
10. A pharmaceutical composition according to claim 9, wherein said agent affecting cytotoxic activity is at least one of radionuclides technicium or iridium, nitroimidazoles and cytokines.
11. Use of a composition consisting essentially of a uniformly dispersed vasoconstrictive drug, present in a therapeutically effective amount, and at least one uniformly dispersed cytotoxic drug present in a therapeutically effective amount, in a non-proteinaceous aqueous medium to treat a neoplastic lesion or surrounding tissue, whereby said drug is retained in the immediate environment avoiding significant levels of the drug at sites distant from the site of application.
12. Use as claimed in claim 11, wherein said vasoconstrictive drug is at least one of the following:
epinephrine, norepinephrine, the catecholamines, epinephrine, isoproterenol, dopamine, ephedrine, phenylephrine, amphetamine, metraminol, methoxamine, ergot alkaloids, ergonovine, methylergonavine, methysergide, ergotamine, an angiotensin, and a prosta-glandin: said vasoconstrictive drug present in a therapeutically effective amount.
13. Use according to claim 11, wherein said cytotoxic drug is at least one of the following: an alkylating agent, enzyme inhibitor, lytic agent, DNA synthesis inhibitor, membrane permeability modifier, DNA
intercalator, or antimetabolite.
14. Use according to claim 11, wherein said cytotoxic drug is at least one of the following: cisplatin, doxorubicin hydrochloride, bleomycin sulfate, fluorouracil, vincristine sulfate, vinblastine sulfate, chlorambucil, melphalan, busulfan, carmustine, lomustine, streptozotocin, thiotepa, dacarbazine, methotrexate, cytarabine, azaribine, mercaptopurine, thioguanine, actinomycin D, plicamycin, mitomycin-C, asparaginase, procarbazine hydrochloride, prednisone , prednisilone, triamcinolone, testosterone, estrogen, insulin, BUdR, nitrosourea, hydroxyurea.
15. Use according to claim 11, wherein said composition in addition consists of an agent affecting tissue architecture and present in a therapeutically effective amount.
16. Use according to claim 15, wherein said agent affecting tissue architecture is at least one of papain, chymopapain, trypsin, amylase, collagenase and chymotrypsin.
17. Use according to claim 11, wherein said composition in addition consists of an agent affecting cellular permeability and present in a therapeutically effective amount.
18. Use according to claim 17, wherein said agent affecting cellular permeability is at least one of detergents amphotericin B and dimethylsulfoxide.
19. Use according to claim 11, wherein an affecting cytotoxic activity which is at least one of radionuclides technicium or iridium, nitroimidazole and cytokines is present in said medium in a therapeutically effective amount.
20. Use according to claim 11, further including the use of radiation or heat, applied in a therapeutically effective amount to the lesion site.
CA000578596A 1987-09-28 1988-09-27 Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas Expired - Fee Related CA1330295C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US101,599 1987-09-28
US07/101,599 US4978332A (en) 1987-09-28 1987-09-28 Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas

Publications (1)

Publication Number Publication Date
CA1330295C true CA1330295C (en) 1994-06-21

Family

ID=22285494

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000578596A Expired - Fee Related CA1330295C (en) 1987-09-28 1988-09-27 Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas

Country Status (5)

Country Link
US (1) US4978332A (en)
EP (1) EP0335966A4 (en)
JP (1) JP2750138B2 (en)
CA (1) CA1330295C (en)
WO (1) WO1989002734A1 (en)

Families Citing this family (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5342613A (en) * 1988-12-27 1994-08-30 Health Research Inc. Pharmaceutical compositions and use thereof in the treatment of psoriasis
US5051257A (en) * 1989-05-09 1991-09-24 Pietronigro Dennis D Antineoplastic solution and method for treating neoplasms
IL95500A (en) * 1989-09-11 1997-03-18 Matrix Pharma ANTI-PROLIFERATIVE COMPOSITIONS CONTAINING TGF-b PROTEIN IN A VISCOUS MATRIX AND THEIR USE
IT1238231B (en) * 1989-12-18 1993-07-12 Consiglio Nazionale Ricerche USE OF IMMUNOMODULANTS AS SYNERGIC AGENTS OF CHEMOTHERAPY IN CANCER THERAPY
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
US5856115A (en) * 1991-05-24 1999-01-05 Fred Hutchinson Cancer Research Center Assay for identification therapeutic agents
US5681812A (en) * 1991-12-10 1997-10-28 Rush Presbyterian-St. Luke's Medical Center Methods and compositions for reducing multidrug resistance
JPH07502274A (en) * 1991-12-10 1995-03-09 ラシユ−プレスビタリアン−セントルークス・メデイカル・センター Methods and compositions for reducing multidrug resistance
EP0562306B1 (en) * 1992-03-06 1995-08-30 Zdzislaw Dr. Fiutowski Pharmaceutical composition having antiviral and antibacterial activity
US5422261A (en) * 1993-04-16 1995-06-06 Baxter International Inc. Composition containing collagenase and chymopapain for hydrolyzing connective tissue to isolate cells
US5626862A (en) 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
CA2224253A1 (en) * 1995-06-09 1996-12-27 Martin J. Macphee Chitin hydrogels, methods of their production and use
US5670358A (en) * 1995-10-19 1997-09-23 Baxter International Inc. Method for inhibiting chymopapain and papain enzyme activity with polysaccharides of animal origin
US5637085A (en) * 1995-11-20 1997-06-10 Cardinale; Robert M. Method of cancer tumor treatment by slow release delivery of 1,2,4-benzotriazine oxides to tumor site
US6010881A (en) 1996-02-20 2000-01-04 Smithkline Beecham Corporation RibG
US5753637A (en) * 1996-10-09 1998-05-19 Ideal Ideas, Inc. Method of treating acne conditions
US5910312A (en) * 1996-10-09 1999-06-08 Ideal Ideas, Inc. Acne treatment composition with vasoconstrictor
US5916574A (en) * 1996-10-09 1999-06-29 Ideal Ideas, Inc. Method of treating natural poison skin conditions
US5919135A (en) * 1997-02-28 1999-07-06 Lemelson; Jerome System and method for treating cellular disorders in a living being
IL132120A0 (en) 1997-04-03 2001-03-19 Guilford Pharm Inc Biodegradable terephthalate polyester-poly (phosphate) polymers compositions articles and methods for making and using the same
WO1999025863A1 (en) * 1997-11-14 1999-05-27 Cedars-Sinai Medical Center Transfection and transfer of male germ cells for generation of transgenic species
US6734338B1 (en) 1997-11-14 2004-05-11 Cedars-Sinai Medical Center Transfection, storage and transfer of male germ cells for generation of transgenic species and genetic therapies
US7294755B1 (en) 1997-11-14 2007-11-13 Cedars-Sinai Medical Center Genetic modification of male germ cells for generation of transgenic species and genetic therapies
EP1107788A1 (en) * 1998-07-23 2001-06-20 Smithkline Beecham Corporation Scrp-5: secreted cysteine rich protein-5
US6419709B1 (en) 1998-10-02 2002-07-16 Guilford Pharmaceuticals, Inc. Biodegradable terephthalate polyester-poly(Phosphite) compositions, articles, and methods of using the same
US6153212A (en) * 1998-10-02 2000-11-28 Guilford Pharmaceuticals Inc. Biodegradable terephthalate polyester-poly (phosphonate) compositions, articles, and methods of using the same
US6350464B1 (en) 1999-01-11 2002-02-26 Guilford Pharmaceuticals, Inc. Methods for treating ovarian cancer, poly (phosphoester) compositions, and biodegradable articles for same
US6537585B1 (en) 1999-03-26 2003-03-25 Guilford Pharmaceuticals, Inc. Methods and compositions for treating solid tumors
US6645947B1 (en) 1999-05-20 2003-11-11 Chitogenics, Inc. Adhesive N, O-carboxymethylchitosan coatings which inhibit attachment of substrate-dependent cells and proteins
US6635677B2 (en) * 1999-08-13 2003-10-21 Case Western Reserve University Methoxyamine combinations in the treatment of cancer
US6428785B1 (en) 1999-10-28 2002-08-06 Immunolytics Inc. Method and composition for treating prostate cancer
US7056697B2 (en) * 2000-10-06 2006-06-06 Affinium Pharmaceuticals, Inc. FabK variant
US7033795B2 (en) * 2000-10-06 2006-04-25 Affinium Pharmaceuticals, Inc. FabK variant
US7048926B2 (en) * 2000-10-06 2006-05-23 Affinium Pharmaceuticals, Inc. Methods of agonizing and antagonizing FabK
WO2003007915A2 (en) * 2001-07-19 2003-01-30 Guilford Pharmaceuticals, Inc. Compositions for treatment of head and neck cancers, and methods of making and using the same
WO2003007914A2 (en) * 2001-07-19 2003-01-30 Guilford Pharmaceuticals, Inc. Biocompatible polymer containing composition for treatment of prostate cancers
WO2003015700A2 (en) * 2001-08-15 2003-02-27 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Novel vasoconstrictor cannabinoid analogs
JPWO2003041740A1 (en) * 2001-11-16 2005-03-03 味の素株式会社 Tumor necrosis agent
DE10229456A1 (en) * 2002-07-01 2004-02-05 Wank, Rudolf, Prof. Dr.med. Treatment of skin tumors and warts, by local application of preparations containing dopamine receptor agonists, especially bromocriptine
US7265097B2 (en) * 2002-08-20 2007-09-04 Chitogenics, Inc. Methods of drug delivery using sulphated chitinous polymers
JP2006500996A (en) * 2002-09-26 2006-01-12 エンドバスキュラー デバイセス インコーポレイテッド Apparatus and method for delivering mitomycin via an eluting biocompatible implantable medical device
US20040153025A1 (en) * 2003-02-03 2004-08-05 Seifert Paul S. Systems and methods of de-endothelialization
US20050170020A1 (en) * 2003-05-29 2005-08-04 Munisekhar Medasani Using organic and / or inorganic potassium and its salts to treat autoimmune and other health disorders and methods of administering the same
WO2010001391A1 (en) * 2008-06-30 2010-01-07 Oron Zachar Dermal application of vasoconstrictors
RU2012120785A (en) 2009-10-22 2013-11-27 ПРОПАНК ПиТиУай ЛТД PHARMACEUTICAL COMPOSITIONS
US20120041397A1 (en) * 2010-08-13 2012-02-16 Elorac, Ltd Method of treatment of premalignant and malignant skin lesions with cytotoxic agents
SG10202011046RA (en) * 2012-09-21 2020-12-30 Intensity Therapeutics Inc Method of treating cancer
NZ744845A (en) 2016-01-29 2023-04-28 Propanc Pty Ltd Cancer treatment
WO2018144094A1 (en) * 2016-11-03 2018-08-09 Health Research, Inc. Methods for improving tumor chemotherapy

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1121054B (en) * 1960-11-23 1962-01-04 Merck Ag E Process for the preparation of a new imidazoline derivative and its acid addition salts
US4177263A (en) * 1972-02-28 1979-12-04 Research Corporation Anti-animal tumor method
US3993754A (en) * 1974-10-09 1976-11-23 The United States Of America As Represented By The United States Energy Research And Development Administration Liposome-encapsulated actinomycin for cancer chemotherapy
US4186183A (en) * 1978-03-29 1980-01-29 The United States Of America As Represented By The Secretary Of The Army Liposome carriers in chemotherapy of leishmaniasis
GB2046092B (en) * 1979-03-05 1983-11-02 Toyama Chemical Co Ltd Pharmaceutical composition containing a lysophospholid and a phospholipid
US4558690A (en) * 1982-01-26 1985-12-17 University Of Scranton Method of administration of chemotherapy to tumors
EP0086627B1 (en) * 1982-02-12 1985-08-28 Unitika Ltd. Anti-cancer device
US4619913A (en) * 1984-05-29 1986-10-28 Matrix Pharmaceuticals, Inc. Treatments employing drug-containing matrices for introduction into cellular lesion areas
US4710493A (en) * 1984-08-14 1987-12-01 Albert Landsberger Therapeutic agent for the use in cancer treatment
DE3432661A1 (en) * 1984-09-05 1986-03-06 Albert Prof. Dr. 6907 Nußloch Landsberger CARCINOM THERAPEUTIC

Also Published As

Publication number Publication date
US4978332A (en) 1990-12-18
EP0335966A1 (en) 1989-10-11
WO1989002734A1 (en) 1989-04-06
EP0335966A4 (en) 1990-02-20
JPH02502015A (en) 1990-07-05
JP2750138B2 (en) 1998-05-13

Similar Documents

Publication Publication Date Title
CA1330295C (en) Treatments employing vasoconstrictive substances in combination with cytotoxic agents for introduction into cellular lesion areas
EP0167263B1 (en) Drug-containing matrices for introduction into cellular lesion areas
EP0221176B1 (en) Pharmaceutical vehicles for recucing transdermal flux
USRE35748E (en) Treatments employing drug containing matrices for introduction into cellular lesion areas
KR20080019228A (en) A carrier comprising one or more di and/or mono-(electron transfer agent) phosphate derivatives or complexes thereof
JP5642892B2 (en) A combination containing macitentan for the treatment of glioblastoma multiforme
US11779644B2 (en) Sensitizing cells to proton radiation
US8293787B2 (en) Gallium compositions for the treatment of liver cancer and methods of use
WO2007140280A1 (en) Anti-cancer composition and method for using the same
JP2004507451A (en) Methods and compositions for the treatment of cancer by administration of an apoptosis-inducing chemotherapeutic agent
US20070248629A1 (en) Pharmaceutical formulations for iontophoretic drug delivery
EP1872798B1 (en) Intestinal absorptive anti-tumor agent
WO2010123547A1 (en) Versatile nanoparticulate biomaterial for controlled delivery and/or containment of therapeutic and diagnostic material
US7608612B2 (en) Radiosensitizer formulations and methods for use
WO2004030661A2 (en) Therapeutic compositions comprising a boron-containing compound
Begg et al. Improvement of combined modality therapy with cisplatin and radiation using intratumoral drug administration in murine tumors
EP0866709B1 (en) 1,2,4-benzotriazine oxides formulations
EP0328389B1 (en) Compostions for treating intracranial tumors
KR20100126453A (en) Combination anti-cancer agents
Kuroda et al. Thermal enhancement of the effect of ifosfamide against a spontaneous murine fibrosarcoma, FSa-II
US20080038376A1 (en) Anti-cancer composition and method for using the same
US20050266076A1 (en) Resorbable delivery systems for the treatment of cancer
WO2003041645A2 (en) Method of vaccinating a human patient to prevent metastatic tumors
CA2494209A1 (en) Radiosensitizer formulations and methods for use
Coderre et al. Boron neutron capture therapy of ocular melanoma and intracranial glioma using p-boronophenylalanine

Legal Events

Date Code Title Description
MKLA Lapsed