CA1329769C - Liposomes with enhanced circulation time - Google Patents

Liposomes with enhanced circulation time

Info

Publication number
CA1329769C
CA1329769C CA000555256A CA555256A CA1329769C CA 1329769 C CA1329769 C CA 1329769C CA 000555256 A CA000555256 A CA 000555256A CA 555256 A CA555256 A CA 555256A CA 1329769 C CA1329769 C CA 1329769C
Authority
CA
Canada
Prior art keywords
liposomes
blood
liposome
res
gml
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
CA000555256A
Other languages
French (fr)
Inventor
Theresa M. Allen
Alberto Gabizon
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.)
Liposome Technology Inc
Original Assignee
Liposome Technology 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
Priority claimed from US06/946,415 external-priority patent/US4837028A/en
Application filed by Liposome Technology Inc filed Critical Liposome Technology Inc
Application granted granted Critical
Publication of CA1329769C publication Critical patent/CA1329769C/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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S436/00Chemistry: analytical and immunological testing
    • Y10S436/829Liposomes, e.g. encapsulation
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]
    • Y10T428/2984Microcapsule with fluid core [includes liposome]

Abstract

ABSTRACT
A composition of liposomes which contain an entrapped pharmaceutical agent and are characterized by (a) liposome sizes predominantly between about 0.07 and 0.5 microns; (b) at least about 50 mole percent of a membrane-rigidifying lipid, such as sphingomyelin or neutral phospholipids with predominantly saturated acyl chains; and (c) between about 5-20 mole percent of a glycolipid selected from the group consisting of ganglioside GM1, saturated phosphatidylinositol, and galactocerebroside sulfate ester.

Description

~ 3~97 6q 1. Field of the Invention The present invention relates to liposome therapeutic compositions, and particularly to liposomal formulations which have enhanced circulation time ;n the bloodstream, when administered intravenouslyO

,~, 2. References ; 15 1. Allen, T.M. (1981) Biochem. Biophys. Acta ;~; 640, 385397.
2. Allen, T.M., and Everest, J. (1983) J.
Pharmacol. Exp. Therap. 226, 539-54~.
3. Altura, B.M. (1980) Adv. Microcirc. 9, 252-294.
4. Alving, C.R. (1984) Biochem. Soc. Trans. 12, `~ 342344.
5. Ashwell, G., and Morell, A.G. (1974) Adv.
'~ Enzymology 41, 99-128.
6. Czop, J.K. (1978) Proc. Natl. Acad. Sci. USA
' 75 3a3l.
, _ 7. Durocher, J~P., et al (1975) Blood 45:11.
8. Ellens, H., et al. (1981) Biochim. Biophys.
Acta 674, 10-18.
9. Gregoriadis, G., and Ryman, B.E. (1972) Eur.
J. Biochem. 24, 485-491.
10. Gregoriadis, G., and Neerunjun, D. (1974) Eur. J. Biochem. 47, 179-185.
11. Gregoriadis, G., and Senior, J. (1980~ FEBS
Lett. 119, 43-46.

:;

.
~ - .
, ~
'' . , ~ ' ~
.

.

-2- 1 32976'3 12. Greenberg, J.P., et al (1979) Blood 53-916.
13. Hakomori, S. (1981) Ann. Rev. Biochem. 50, 733-764.
14. Hwang, K.J., et al (1980) Proc~ Natl. Acad.
Sci. USA 77:4030.
15. Jonah, M.M., et al. (1975) Biochem. Biophys.
Acta 401, 336-348.
16. Juliano, R.L., and 5tamp, D. (1975) ~iochem.
Biophys. Res. Commu~. 63, 651-658O
17. Karlsson, K.A. (1982) In. Biological Mem-branes, vol. 4, D. Chapman (ed.) Academic Press, N.Y., pp. 1-74.
18. Kimelberg, H.K., et al. (1976) Gancer Res.
36, 2949-2957.
19 Lee, K.C., et al, J. Immunology 125:86 (1980).
20. Lopez-Berestein~ G., et al. (1984) Cancer Res. 44, 375-378.
21. Okada, N. (1982) Nature 2g9:261.
22. Poznansky, M.J., and Juliano, R.L. (1984) Pharmacol. Rev. 36, 277-336.
23. Richardson, V.J., et al. (1979) Br. J. Cancer 40, 3543.
24. Saba, T.M. (1970) Arch. Intern. Med. 126, 1031~1052.
25. Schaver, R. (1982) Adv. Carbohydrate Chem.
Biochem. 40:131.
: 26. Scherphof, T., et al. (1978) Biochim.
Biophys. Acta 542, 296-307.
27. Senior, J., and Gregoriadis, G. (1382) FEBS
Lett. 145, 109-114.
28. Senior, J., et al. (1985) Biochim. Biophys.
Acta 839, 1-8.
29. $zoka, F,, Jr., et al (1978) Proc. Natl.
Acad. Sci. USA 75:4194.

~' A

.. . .
~, . . ... . . . . .

,. . . . . ... . . . ....
. ... ~ . . .. . ....
- . , ....... , ^ "
". .. ;., . ~ , ...... .

: ~ i ~3~ 1 329769 30. Szoka, F., Jr., et al (1980) Ann. Rev.
- Biophys. Bioeng. 9:467.
31. Woodruff, J.J., et al (1969) J. Exp. Med.
129~51.

3. Backqround_of the Invention Liposome delivery systems have been proposed for a ~ariety of drugs. ~or use in drug delivery via the bloodstream, liposomes have the potential of provid-ing a controlled "depot" release of a liposome-entrapped drug over an ~xtended time period, and of reducing toxic side effects of the drug, by limiting the concentration of free drug in the bloodstream. Liposome/drug composi-tions can also increase the convenience of therapy by allowing higher dru~ dosage and less frequent drug a~ministration. Liposome drug delivery systems are reviewed generally in Poznansky et al, One limitation-of intravenous liposome drug delivery which has been recognized for many years is the rapid uptake of blood-circulating liposomes by the mono-nuclear phagocytic system (MPi'i), also referred to as thereticuloendothelial system (Rl~.S). This system, which consists of the circulating macrophages and the fixed macrophages of the liver (~upffer cells~, spleen, lungs, and bone marrow, removes foreign particulate matter, ;- including liposomes, from blood circulation with a half e in the order of minutes (Saba~. Lipo~omes, one of the most ~tensively investigated par~iculate dru~ car-riers, ~re removed from cireulation primarily by Kupffer cells of the liver and to a lesser extent by o~her mac-~; rophage populations.
A Yariety of studies on factors which effect . liposome uptake.byi~he RES have been reported. Ear~y experiments, using heterogeneous preparations of ,~
~' . _--. ~ . . .

. .

4~ 1 32 ~7 6q multilamellar liposomes (MLV) containing phosphatidylcholine (PC) and cholesterol (CH) as their principal lipid cons~ituents, demonstrated that these liposomes are rapidly removed from circulation by uptake into liver and spleen in a biphasic process with an ini-tial rapid uptake followed by a slow phase of uptake (Gregoriadis, 1974; Jonah; Gregor;adis, 1972 Juliano).
Half-life for removal of MLV from circulation was on the order of 5-15 min. following intravenous (IV) injection.
Negatively charged liposomes are removed more rapidly frsm circulation than neutral or positively charged liposomes. Small unilamellar liposomes (SUV) are cleared with half-lives approximately three-to four-fold slower than MLV liposomes (Juliano; Allen, 1983). Uptake of liposomes by liver and spleen occurs at similar rates in several species, including mouse, rat, monkey, and human (Gregoriadis, 1974; Jonah; Rimelberg, 1976; Juliano;
Richardson; Lopez-Berestein).
Liposomes which are capable of evading the RES
would have two important benefits. One is the increased liposome circulation time in the blood, which would both increase the pharmacokinetic benefits of slow drug release in the bloodstream, and also provide greater opportunity for tissue targeting where the liver, spleen, and lungs are not involved. The second benefit is decreased liposome loading of the ~ES. In addition to the role of the RES in removing foreign particles, the RES is involved in several other functio~s, includ-ing host defense against pathogenic microorganisms, par-asites, and tum~r cells, host responses to endotoxins and hemorragic shock, drug response, and responses tocirculating ~mmune complexes (Saba, Altura). It is --important~; there~ere, in liposome administration via the , :.-, , ..

-5- 1 3 2 9 7 6q bloodstream, to avoid compromising the RES seriously, by massive short-term or accumulated liposome uptake.
One approach which has been proposed is to increase liposome circulation time by increasing liposome stability in serum. This approach is based on studies by one of the inventors and others which have shown that factors which decrease leakage of liposome contents in plasma also decrease the rate of uptake of liposomes by the ~ES ~Allen, 1983; Gregoriadis, 1980;
Allen, 1981; Senior, 1982). The most important factor contributing to this effect appears to be bilayer rigid-ity, which renders the liposomes more resistant to the destabilizing effects of serum components, in particular high density lipoproteins (Allen, 1981; Scherphof).
Thus, inclusion of cholesterol in the liposomal bilayer can reduce the rate of uptake by the RES (Gregoriadis, 1930; Hwang; Patel, 1983; Senior, 1985), and solid liposomes such as those composed o~
distearoylphosphatidylcholine (DSPCi or containing large amounts of sphingomyelin (SM) show decreased rate and ext~nt of uptake into liver IAllen, 1983; Ellens;
Senior, 1982; Hwang).
However, this approach appears to have a lim-ited potential for increasing liposome circulation times in the bloodstream. Studies carried out in support of the present invention, and reported below, indicated that 0.1-0.2 micron liposomes containing optimal membrane-rigidifying liposome formulation components are predominantly localized in the RES two hours after intra-venous liposome administration. Although longer circulationtimes are achieved with small unilamellar vesicles or ; SUVs (having a size range between about 0.03 and 0.08 r,: mi~ons)~ W ~ are generally le~s useful in drug de~iv-ery due to their smaller drug-carrying capacity, and .

.
"

' , : .
, . . .

. . . .. . .
. , ~ , ~ , -6- 7 3 ~7 6 ~

their greater instability, which can lead to rapid release of liposome-associated drug, and to liposome fusion events that produce large and heterogeneous-size liposomes.
Several investigators, including the appli-cants, have also explored the possibility of increasing liposome circulation times by designing the liposome surface to mimic that of red blood cells. ~he role of cell surface carbohydrates in cellular recognition phe-nomena is widely appreciated (Ashwell, Hakomori, Karlsson). The chemistry, metabolism, and biological functions of sialic acid have been reviewed (Schauer).
Surface sialic acid, which is carried by gangliosides, and glycoproteins such as glycophorin, plays an impor-tant role in the survival of erythrocytes, thrombocytes, and lymphocytes in circulation. Enzymatic removal ofsialic acid, which exposes terminal galactose residues, results in rapid removal of erythrocytes from circula-tion, and uptake into Kupffer cells of the liver ~Durocher). Desialylation of thrombocytes (Greenberg) and lymphocytes (Woodruff~ also results in their rapid i removal by the liver.
I Although desialylated erythrocytes will bind to Kupffer cells or peritoneal macrophages in vitro in the a~sence of serum, serum must be added in ~rder for significant phagocytosis to occur. The nature of the serum components mediating endocytosis is speculative, but i~munoglobin and complement (C3b) are thought to be involvsd. Czop et al. have shown that sheep erythrocytes, which are not normally phagocytosed by human monocytes, will bind C3B and be phagocytosed upon desialylation.
-;; Okada et al. have demostrated "i;,~hat sia~lyglyco~ipids on liposome membranes restrict activation of the alternative complement pathway and /

.~, ~. .

, . . . .,- . , :: . :::
, ~, ; . . ,., ,, . :
. . , ., ~. :

~7~ 1 329769 that removal of the terminal sialic acid from the glycolipids abolishes this restricting capacity and results in activation of the alternative complement pathway. Sialic acid, therefore, may be functioning as a non-recognition molecule on cell membranes partly through its ability to prevent binding of C3b, thus pre-venting phagocytosis via the alternative complement pathway. Other immune factors may also be involved in liposome phagocytosis. Alving has reported ~hat 50% of the test sera from individual humans contain naturally occurring "anti-liposome" antibodies which mediated complement-dependent immune damage to liposomes.
The observations reported above suggest that surface sialic acid, and/or other red-cell surface agents, inçorporated into liposomes, for example, in the ! form sf ganglioside or glycophorin, may lead to increased circulation half-lives of liposomes. This approach is described, for example, in U.S. Patent No.
4, 501, 728 for "Masking of Liposomes from RES Recogni-tion", although this patent does not disclose whether significant RES masking is actually achieved by coating liposomes with sialic acid.
In fact, experiments conducted in support of the present application indicate that sialic acid, in the form of gangliosides, has a limited ability to extend circulation half lives in vivo in liposomes which are predominantly composed of conventional liposomal lipid~, such as egg phosphatidylcholine (egg PC) or egg PC:cholesterol mixtures. In vivo uptake studies on PC:cholesterol:ganglioside liposomes (0.2 microns or i, smaller~ indicate that the injected liposomes are local-¦ ized predominantly in the RES two hours post ~ administration. ~

s~

... .. : :::, s ! . ., :" ' ' '' ~, ' ~ ' .: . .

-8- 1 32976~

In summary, several approaches for achieving enhanced liposome circulation times in the bloodstream hav~ been proposed. Heretofore, however, the approaches have produced quite limited improvements in blood circu-lation times, particularly in liposomes in the 0.07-0.2 micron size range which are generally most desirable for parenteral drug compositions.

4. Summary of the Invention It is therefore one general object of the pre-sent invention to provide an improved liposome composi-tion which gives significantly improved blood circula-tion times.
A more specific object of the invention is to provide such a composition in which liposomes are pre-dominantly localized in the bloodstream, rather than inthe liver and spleen, several hours a~ter liposome administration.
Yet another object of ~he inven~ion is to pro-vide such a composition conta;ning liposomes which are in a selected size range between about 0.08-0.2 microns.
Providing a method for significantly extending the lifetime of drug-carrying liposomes in the bloodstream, for improved druq delivery and/or drug tar-geting-in tumor treatment is still another object of the invention.
The invention includes a liposome composition which is designed for enhanced circulation in the bloodstream. The liposomes in the composition are char-acterized ~y~ (a) liposome sizes in a selected size range between 0.07 and 0.4 microns; (b) substantially homogenous-phase liposome bylayers composed of at least 50 mole percent of a membrane-rigidifying lipid, and (c) between 5-20 mole percent of a .

~.: '' .' ;' ' '":'''' :; ;,. ..
. ~. , ~ .

:. , ~ , , .

_g_ 1 32976q glycolipid component selected from the group consisting of ganglioside ~Ml, hydroqenated phosphatidylinositol, and sulfatide, i.eO, sulfate es~ers of lactocerebrocidesmonogalactosyl. ThP lipophilic moiety of the glycol;pid serves to anchor the component ;n the liposome ~ilayers, without phase separation therein.
The liposomes show enhanced tissue distribu~
tion, as measured by the ratio of liposomal marker pre-sent in the blood to the combined amount of marker in the liver and spleen, when measured 2, 4, and 24 ~ours after intravenous liposome administration. Preferably, blood/RES ratio, when measured 2 hours after administra-tion, is substantially greater than the sum of the blood/RES ratios obtained with similarly constructed liposome compositions containing in one case, at least about 50 mole percent of the membrane rigidifying lipid, but.no~ the glycolipid, and in the other case, between ; 5-20 mole percent of the glycolipid, but not the membrane-rigidifying lipid.
In one preferred embodiment, the membrane-rigidifying lipid is a combination of sphingomyelin and a neutral phospholipid, such as phosphatidylcholine (PC), in a preferred molar ratio of between 2 1 and 4:1. In another embodiment, ~he lipid is a saturated PC, with or without cholesterol.
The method of the invention is designed for extending the bloodstream lifetime of drug-containing liposomes which are administered intravenously in a 5us-pension of liposomes whose sizes are in a selected size 30 range between about 0.07-0.4 microns. The method includes preparing the liposomes to contain (a) at least a~o~t ! ~O--mole percent of a membrane-rigidifying lipid in :the:l~posome bilayers, which are subs~antially ;
phase-homogeneous, and (b) between 5-20 mole ~ .

~.

C,t !
-lo- 1 329769 percent of a glycolipid component selected from the group consisting of ganglioside GM1, hydrogena~ed phosphatidylinositol, and sulfatide.
The method gives significantly enhanced drug uptake in tumors, when administered intravenously to a tumor-bearing subject. The method is therefore useful in administering an anti-tumor compound for the treat-ment of the tumor by intravenous administration.
These and other objects and features of the inven~ion will become more fully apparent when the fol-lowing detailed description of the invention is read in conjunction with the accompanying figures.

Brief Description of the Fiaures Figure 1 shows the time course of decrease of blood/RES ratios in a test subject injected IV with ~1) liposomes containing ganqlioside GMl, but not SM (solid circles) and (2) with liposomes containing both GMl and SM (open circles);
Figures 2A and 2B show the effect of increas-ing molar amounts of GMl on percent injected liposome marker in blood (circles), liver (triangles), and spleen (squares) two hours post injection, in liposomes con-taining either PC:CH, 2:1 (A) or SM:PC, ~:1 (B) and Figures 3A and 3B show linear regression plots of different liposome formulations for (A) liposomal marker in the RES versus the marker in the bloodstream, and (B~ liposomal marker in the J6456 tumor (solid tri-angles) and B-16 tumor (solid circles~ versus amount in the bloodstream; and Figure 4 shows the glycolipids ganglioside GM
(A), hydrogenated phosphatidylinositol (B~ and monogalact~syl stearate sulfatide ~G) which are sompo-nents of the liposome compositions of the invention.

~, ..
.....
` ''' .

-;. .
,. , ; ,", - ,,, ~.. . ' ' ' .~. . ;: ,. . . . .

.

(-~

Detailed Description of the Invention The liposome composition of the invention is desiyned for delivering a drug or other agent, such as nutritional supplements, vitamins, or chelated metal, to a subject via the bloodstream, and for relatively slow uptake of the liposomes by the RES, allowing the drug or agent to be released from the liposomes into the bloodstream over an extended period of several hours or more. Alternatively, the composition is designed, by appropriate surface modification of the liposomes, for targeting via the bloodstream to non-RES target tissues, to allow the drug or agent to concentrate in the immedi-ate region of the target tissue.
15Section IA below describes the general method used to evaluate liposome uptake by the RES in vivo, section IBj the combination of liposome components which have been found, according to one aspect of the inven-tion, to give high blood circulation times for intrave-~, 20 nously injected liposomes, and section IC, methods for preparing, sizing, and sterilizing drug-containing liposomes designed for intravenous administration. The I utility of the liposome composition, in drug delivery I and drug targeting, is discus.sed in Section II.
I. PreDarinq the LiPosomal ComPosition I A. Measuring liposome uptake by the RES in vivo The method used for evaluating liposome circu-lation time in vivo measures the distribution of intra-venously injected liposomes in the bloodstream and the .p~imary-organs:o~ the RES at se~ected times after injec-.i . tion. In the standardized model which is used, RES
, uptake is measured by the ratio of total liposomes in :~, ~. _ j . ,......... ,. : :, , ,. , :

:,. - ., . : , , .
: . .~,,.. , .,... , , ~
, : ~ , . .. , , . - :
:: : - : ; . :: ,: , . . . . . :

, . , .: ;. ... .. . .
. : ,. . .
. : i . ,~: .

(~ ( ) -12- 1 3 2 97 6~

the bloodstream to total liposomes in the liver and spleen, the principal organs of the RES. In practice, age and sex matched mice are injected intravenously (IV) through the tail vein with a radiolabeled liposome com-position, and each time point is determined by measurin~total blood and com~ined liver and spleen radiolabel counts, and in many studies, complete dissection, weigh-ing, and radioactivity determinations of all body parts was done. Total blood counts are calculated by assuming that the total blood volume makes up 7% of the animal's body weight. Experimental methods are detailed in Exam-ple 2.
Since the liver and spleen account for nearly 100 % of the initial uptake of liposomes by the RES, the blood/RES ratio just described provides a good approxi-mation of the extent of uptake from the blood to the RESin v_o. For example, a ratio of about 1 or greater indicates a predominance of injected liposomes remaining in.the bloodstream, and a ratio below about 1, a predom-inance of liposomes in the RES. For most of the lipid compositions of interest, blood/RES ratios were calcu-lated at 2, 4, and 24 hours.
The data obtained with the model animal system can be reasonably extrapolated to h~mans and veterinary animals of interest. This is because, as mentioned above, uptake of liposomes by liver and spleen has been found to occur at similar rates in several mammalian species, ineluding mouse, rat monkey, and human (Gregoriadis, 1974; Jonah; Kimelberg, 1976; Juliano;
3~ Richardson; Lopez-Berestein). This result likely reflects the fact that the biochemical factors which appear to be most important in liposome uptake by the . ~RES-~including opsinization by.serum lipoproteins, siæe-dependent uptake effects, and cell shielding by ~ ., .
~ ~ ' ' ' ' , ( l surface moieties are common featur~s of all mammalian species which have been examined.

B. Lipid ComPonents The lipid components used in forming the liposomes of the present invention are selected to meet three important criteria:
First, a major portion of ~he lipids, i.e., more than 50 mole percent, are neutral membrane-rigidifying components, by which is meant uncharged lipid components which produce relatively rigid, close-packed lipid bilayer structures. Typi-cally, the lipids are predominantly saturated lipids whose phase transition temperature (Tc) is above about 25C, and preferably between about 30~C and 50C. Pre-ferred membrane-rigidifying lipids include SM (Tc about 30CC) and neutral phospholipids, such as PC whose acyl chains are predominantly saturated. One saturated PC
which has been investigated ~xtensively herein is distearoyl PC ~DSPC) whose Tc is about 50C.
Secondly, and according to an important fea-ture of the invention, the lipid components must contain a negatively charged glycolipid which has a single nega-tive charge which appears to be partially shielded, i.e., inaccessible ~o certain types of charge-charge bindin~ -interactionr, on the outer surface of the liposomes. To date, three glycolipids which have this feature, as evidenced by hi~h blood/RES ratios achieved with the liposome formulation have been identified~
These are yanglioside GMl, hydroqenated phophatidylinositol (HPI), and sul~atides (~), i.e., sulfate esters of gala~tocerebro~ides. These three Fompounds are sh~wn in~ Figures lA-lCJ respectively, where the R
group in the GMl and sulfate compounds are long-chain '`''"`~ .
~' " .
.. ~ . , .. , ; .~ .
.; ~ . . . - , ,.:... i hydrocarbons. The particular sulfatide shown in th~
figure is cerebronic acid.
Thirdly, the above lipid components must pro-duce a substantially homogeneous-phase bilayer struc-ture, by which is meant that the critical membranerigidifying and negatively charged lipid components exist in a single phase, as opposed to discrete phases which are physically separate. As will be seen, this requirement both enhances blood/RES values achievable with the liposomes, and also minimizes rapid liposome breakdown and leakage in the bloodstream.
The importance of membrane-rigidying lipid components in combination with one of the selected glycolipids can be appreciated from blood/RES data pre-sented in ~xamples 3, 4, 8 and 9. Table 1 in Example 3 shows several liposome compositions, and correspondingblood/R~S values measured 2 hours post injection. A
comparison of compositions 3 and 4 in the table shows the large increase in blood/RES values which are obtained by addition of cholesterol to liposomes con-taining egg PC and GMl. Cholesterol is known to have a rigidifying or "packing" effect on relatively fluid lip-ids, such as egg PC, which has a Tc of about 0C. More importantly, for purposes of the present invention, a comparison of compositions 3 and 4 with compositions 12 and 13 indicate that GMl in combination with SM gives much higher blood/RES values than the ganglioside in combination with PC. Here it is noted, with reference to composit;ons 17 and 18, that cholesterol decreases blood/RES ratios in SM:GMl formulations, presumably because of the known fluidizing effect of cholesterol in some.rigi~-iipid compositions. -. The effect of subsituting DSPC, i.e., a saturated-chain PC, for egg PC in some of the Table 1 ~n - .

. . .

-15- ~ 3~97 69 formulations is seen in Table 2 from Example 4. Clearly DSPC and GMl together effec~ively increase blood/RES
values, an effect which is enhanced still further by addition of SM to the compositions, with highest ratios being observed with the highest relative amounts of SM:PC. Note that cholesterol appears to have less of a fluidizing effect, as evidenced by blood/RES ratios, in the presence of DSPC.
The effect of saturated lipids on blood/RES
values achievable with HPI is seen in Table 5 and 6, where compositions containing PI (unsaturated PI) in P~:CX and HPI in unsaturated PC (DOPC) CH are compared with compositions containing HPI:DSPC:CH. The blood/RES
ratio of the saturated composition is substantially higher than that of the unsaturated compositions (either unsaturated PI or PC). Note also that the saturated composition is more stable in the bloodstream than the two compositions containing unsaturated PI or PC, as evidenced by the greater total recovery after 24 hours.
The requirement for saturated PI, e.g., HPI, is to pre-vent phase separation of the PI component in therigid-lipid bilayer structure.
The effect of saturated lipids on ~he blood/RES ratio of sulfatide containing liposomes can be appreciated from a comparison of the blood/RES data in Ta~le 6~ ~s seen, ~he blood/RES ratio at 24 hours increases from about 15 fold when SULF is used in combi-nation with saturated lipid components. As defined herein, sulfatide includes ios Example 8 examines blood/RES ratios in several liposome compositions 4 hours post injection. The data here shows that HPI, in the presence of saturated phospholipid (DSPC) and cholesterol, gives a blood/RES

.

..... ~
. : ~

value which is about three times greater than that of liposomes composed of unsaturated PI and PC lipids and cholesterol.

Table 6 from Example 9 shows blood/RES values for a number of liposome compositions, as measured 24 hours post injection. The compositions are arranged according to increasing blood/RES values.

It is apparent first that blood/RES values have declined significantly between 4 and 24 hours, but that the best formulations sitll show relatively good blood/RES
values, e.g., 0.4 or higher, after 24 hours. These formulations include HPI, and GMl in combination with saturated PC (DSPC) plus cholesterol, in the presence or absence of SM.

The liposomes, the above-discussed data, and particularly the comparative data from Table 6, indicate that both GMl and HPI, are effective, in combination with membrane-rigidifying components, in producing high blood/RES
ratios. The ability of GMl, in comhination with membrane-rigidifying components, to produce high blood/RES
ratios has been disclosed in parent co-pending application, now UOS. patent No. 4,598,051. The present invention shows that a similar effect of HPI in liposomes formed predominantly of rigid-lipid membrane components.

'~,~

, .
.; , ~ , , ~ 1 329769 In Table 5, it is seen that GMl, SULF, and HPI all give comparable blood/RES values (about 1) in PC:CH liposome, whereas GMl and HPI (SULF was not tested) gave highest values with rigid-lipid components.
Figures 2A and 2B are plots of blood/RES values as a function of GMl mole ratio in two different liposome formulations.
The first, shown in Figure 2A, is a PC:CH formulation twhich gives suboptimal blood/RES ratios), and the second, an SM:PC formulation. As discussed in Example 6, only the latter formulation shows a strong GMl is be-tween 5-15 mole percent.
The effect of high glycolipid concentration on blood/RES ratios is seen in Example 10, which examines 4 and 24 hour blood/RES ratios for liposomes containing DSPC:CH or PC:CH and increasing molar concentrations of HPI.
Molar ratios of HPI about about 25% substantially eliminated the enhanced blood/RES values seen at concentrations of about 16 mole percent or below.

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

The requirem~nt that the lipid components form a homogeneous-phase bilayer is aimedr in part, at ensuring good liposome stability in the bloodstream over a period of 24 hours or longer.

Bilayer phase homogeneity should also contribute to improved blood/RES values, since liposome instability would be expected to enhance RES uptake.

This may explain, for example, why the SM:GMl formulation has a relatively low blood/RES ratio after 24 hours (Table 6), yet gives a very high ratio after 2 hours (Table 1)~

The data from both tables indicate that this formulation is quite unstable and/or leaky in the bloodstream, as detailed in Example 9.

Phase inhomogeneity may also explain why PI is less effective than HPI in combinat:ion with DSPC:CH mixtures in enhancing blood/RES ratios, since HPI would be expected to be more phase-compatible (more similar Tc) with saturated PC.

In addition to the membrane-rigidifying agents and gangliosides required in the liposome composition, the liposomes may be formulated to include other neutral vesicle-forming lipids which do not significantly compromise the RES-evasion properties of the liposomes.

.
. ' ' ; ', ' :: ' ' .

--lg--An obvious example is choles~erol which is used in many of the above formulations Above, at a mole ratio of about 30%.

The liposomes may also include protective agents such alpha-tocopherol, or other free-radical inhibitors, to minimize oxidative damage to the liposomes and/or entrapped drug carried in the liposomes.

, ~ .

i, ,~
,~

,:: . .-.. .. ; . -.
: . ~ - : :: :: . - i C. Preparing the Liposome Composition The liposomes may be prepared by a variety of techniques, such as those detailed in Szoka et al, 1980.
One preferred method for preparing drug-containing liposomes is the reverse phase evaporation method described by Szoka et al and in U.S. Patent No.
4,235,871. In this method, a solution of liposome-forming lipids is mixed with a smaller volume of an aqueous medium, and the mixture is dispersed to form a water-in-oil emulsion. The drug or other pharmaceutical agent to be delivered is added either to the lipid solu-tion, in the case of a lipophilic drug, or to the aque-ous medium, in the case of a water-soluble drug. Here it is noted that all lipid and aqueous components should preferably be sterile and pyrogen free. After removing the lipid solvent by evaporation, the resulting gel is converted to liposomes, with an encapsulation effi-ciency, for a water-soluble drug, of up to 50%. The reverse phase evaporation vesicles (REVs) have typical average sizes between ahout 2-4 microns and are predomi-nantly oligolamellar, that is, contain one or a few lipid bilayer shells. The method is detailed in Example lA.
The REVs are readily sized, as discussed below, by extrusion to give oligolamellar vesicles hav-ing a maximum selected size preferably between about 0.08 to 0.4 microns. Experiments conducted in support of the present invention indicate that sized oligolamellar vesicles of this type show substantially higher blood/RES ratios than similar sized multilamellar vesicles (MLVs), and that smaller REVs, e.g., 0.16-0.17 micron sizes, give higher ratios than larger REVs, e.g., 0-~4 microns. Another advantage of REVs is the high ratio of encapsulated drug to lipid which is possible, ~ , ' ~ - , .
, - ~ . . , ..
.. . . .
, ; , ,~. . .

-21- l 32~769 allowing greater drug doses to be administered in a given lipid dose.
; MLVs, where desired, can be formed by simple lipid-film hydration techniques. In this procedure, a mixture of liposome-forming lipids of the type detailed above dissolved in a suitable solvent is evaporated in a vessel to form a thin film, which is then covered by an aqueous medium. The lipid film hydrates to form MLVs, typically with sizes between about 0.1 to 10 microns.
These vesicles, when unsized, show relatively poor blood/RES ratios, as seen in Table 9, for the unextruded MLV composition. Typically, MLVs are sized down to a desired size range of 0.5 or less, and preferably between about 0.07 and 0.12 microns by extrusion, as detailed below.
One effective sizing method for REVs and MLVs involves extruding an aqueous suspension of the liposomes throu~h a polycarbonate membrane having a selected uniform pore size, typically 0.05, 0.08, 0.1, 0.2, or 0.4 microns (Szoka). The pore size of the mem-brane corresponds roughly to the largest sizes of ' liposomes produced by extrusion through that membrane, particularly where the preparation is extruded two or more times through the same membrane. This method of liposome sizing is used in preparing homogeneous-size ~EV and MLV compositions described in the examples below. A more recent method involves extrusion through an asymmetric ceramic filter.
` Alternatively, the REV or MLV preparations can be treated to produce small unilamellar vesicles (Suvs) which are characterized by sizes in the 0.04-0.08 micron range. However, as indicated above, SUVs have a . , , . , ~

~,.- . . . . . .
. - : . . . , , ~:

- , . . . .

relatively small internal volume, for delivery of water-soluble drugs, and they tend to fuse to form larger het-erogeneous size liposomes with heterodisperse drug leak-age and RES uptake characteristics, and are leakier than REVs or MLVs. SUVs can be produced re~dily by homoge-nizing or sonicating REVs or MLVs, as described in Exam-ple lC.
After final sizing, the liposomes can be treated, if necessary, to remove free (non-entrapped) drug. Conventional separation techniques, such as centrifugation, diafiltration, and molecular-sieve chromatography are suitable. The composition can be sterilized by filtration through a conventional 0.45 micron d~epth filter.

II. UtilitY
The significantly increased circulation half life of liposomes constructed as above can be exploited in two general types of therapeutic or diagnostic liposome compositions. The first composition is designed for sustained release of a liposome-associated agent into the bloodstream by circulating liposomes. As seen above, liposomes constructed according to the invention can be maintained prledominantly in the bloodstream up to 24 hours, and therefore sustained released of the drug at physiologically effective levels for up to about 1 day or more can be achieved.
One measure of increased dru~ availability in the bloodstream is the increased area under the curve (AUC) seen with high blood/R~S liposomes. The AUC mea-surement is made, as described in Example 9, by measur-inq ~iposome mar~e~ levels ~ver a-a 24 hour period, for - both blood and liver:~levels. iThe ratio o~ these AUC
values then shows-the extent to which to~al liposome B

.. . . ;
.
. . . .. . . .
. .. . . .. . . ...
. .

( .i availability has been shifted from the liver to the bloodstream. Table 7 in Example 7 demonstrates that high blood/RES values are correla~ed with high AUC
ratios.
The extended l;fetime of the liposomes in the bloodstream also makes it possible for a significant fraction of the injected liposomes to reach the target site before being removed from the bloodstream by the i RES. In particular, it is desired to target tumor tis-sue for drug treatment by intravenous administration to a tumor-bearing subject.
- The use of the liposome composition of the invention for targeting animal tumors is detailed in Examples 11-13. Briefly summarizing the results, liposomes with increased blood/RES ratios produced a 10-30 fold enhancement in tumor uptake over free drug.
Tumor uptake peaked at 24 hours post administration, although high levels of drug in the tumor were seen between 4 and 48 hours post administration. Details of the treatment methods are given in the examples.
A variety of drugs or other pharmacologically active agents are suitable for delivery by ~he liposome composition. One general class of drugs include water-soluble, liposome-permeable compounds w~ich are charac-terized by a tendency to partition preferentially into the aqueous compartments of the liposome suspension, andto equilibrate, over time, between the inner liposomal spaces and outer bulk phase of the suspension. Repre-sentative drugs in this class include terbutaline, 3~ albuterol,jatropine methyl nitrate, cromolyn sodium, pxopranolol, flunisolide, ibuprofen, gentamicin, tobarmycin, pentamidine, penicillin, theophylline, bleomycin, etoposide, captopril, n-acetyl cysteine, verapamil, vitamins, and radio-opaque and : , , . : ,, - . .

, . .

1 32~76q particle-emitter agents, such as chelated metals. Because of the tendency of these agents to equilibrate with the aqueous composition of the medium, it is preferred to store the liposome composition in lyophilized form, with rehydration shortly before administration. Alternatively, the composition may be prepared in concentrated form, and diluted shortly before administration.
A second yeneral class of drugs are those which are water-soluble, but liposome-impermeable. For the most part, these are peptide or protein molecules, such as peptide hormones, enzymes, enzyme inhibitors, apolipoproteins, and higher molecular weight carbohydrates are characterized by long term stability of encapsulation. Representative compounds in this class include calcitonin, atriopeptin, *a*-l antitrypsin (protease inhibitor), interferon, oxytocin, vasopressin, insulin, interleukin-2, superoxide dismutase, tissue plasminogen activator (TPA), plasma factor 8, epidermal growth factor, t~lmor necrosis factor, lung surfactant protein, interferon~ lipocortin, *a*-interferon and erythropoetin.
A third class of drugs are liphilic molecules which tend to partition into the lipid bilayer phase of the liposomes, and which are therefore associated with ~he liposomes predominantly in a membrane-entrapped form. The drugs in this class are defined by an oil/water partition coefficient, as measured in a standard oil/water mixture such as octanol/water, of greater than 1 and preferably greater than about 5. Representative drugs include prostaglandins, amphotericin B, progesterone, isosorbide : . : .

.; , . .

~1 dinitrate, testosterone, nitroglycerin, estradiol, doxorubicin, beclomethasone and esters, vitamin E, cor-tisone, dexamethason~ and esters, and betamethasone valerate.
For sustained drug-release via the bloodstream, the liposome composition is administered intravenously in an amount which provides a suitable drug dosage over the expected delivery time, typically 12-24 hours. The injection may be given as a single bolus or slowly by i.v. drip, to allow gradual dispersal of the liposomes from the site of injection.
Where it is desired to target the liposomes to a selected non-RES tissue site, the liposomes are preferably designed for surface recognition of target-site molecules. For example, in the case of targetin~to a solid tumor, the liposomes may be prepared with surface-bound tumor recognition molecules, such as ~nti-bodies directed against tumor-specific antigens. Methods for coupling molecules of this type are wellknown to those in the fieldO These methods generally involve incorporation into the liposomes of lipid components, such as phosphatidylethanolamine, which can be activated for attachment of surface agents, or derivatized lipophilic compounds, such as lipid derivatized.
bleomycin.
In one particular liposome composition which is useful for radioimaging of solid tumor regions, the ; liposomes are prepared with encapsulated radio-opaque or particlc-emission metal, typically in a chelated form which substantially prevents permeation Shrough the liposome bilayer, and carrying surface-bound bleomycin molecules~ for preferential liposome attachment to tumor si~esb "~
The following examples illustrate methods of preparing liposomes with enhanced circulation times, and for accessing circulation times in vivo and in vitro.
~ The examples are intended to illustrate specific : 5 liposome compositions and methods of the invention, but are in no way intended to limit the scope thereof.

. Ceramides (CER), cholesterol (CH), monogalactosyl-sterate sulfatides (SULF), g~lactocerebrosides (GAL), glucocerebrosides (GLU), and lactosylceremide (LAC) were obtained from Sigma (St.
Louis, MO). Sphingomyelin (SM), egg phosphatidyl-choline ~lecithin) (PC), phosphatidylinositol (PI), hydrogenated phosphatidylinositol (HPI), phosphatidyl-serine ~PS), phosphatidylglycerol ~PG), phosphatidic acid ~PA), phosphatidylethanolamine ~PE), dipalmitoyl-phospha.tidyl glycerol (DPPG), dipalmitoyl PC ~DPPC), dioleyl PC-~DOPC). and distearoyl PC ~DSPC) were obtained ~ from;Avanti Polar Lipids ~Bi.rmingham, AL). Globosides ~GLOB), digalactosyl diglyceride (DGDG), monosialoganglioside (GMl), ganglioside GM2 (GM2), ganglioside GM3 (GM3), trisialoganglioside (GTlb), and disialo~anglioside (GDla) were obtained from 5upelco (Bellefonte, PA).
[125I]-tyraminyl-inulin was made according to published procedures. 67Gallium-8-hydroxyquinoline was supplied by NEN Neoscan (Boston, MA); doxorubicin (adriamycin), from Adria (Columbus, OH), and bleomycin, ~ from Bristol Myers (Syracuse, NY).
: 30 . i ' r~

.. , . ` t : :
~-: ' ' . :' ~` .; , ..
-27- 1 32976~

Example 1 Preparation of REVs, MLVs and SUVs This example describes the preparation of reverse phase evaporation vesicles (REVs), multilamellar vesicles (MLVs) and small unilamellar vesicles ~SUVs).

A. Sized REYs A total of 50 mg of the selected lipid compo-nents, in the mole ratios indicated in the examples below, were dissolved in 5 ml of diethyl ether. An aqueous buffer containing 13 mM phosphate, 140 mM NaCl, pH 7.4 was added to the organic solvent to a final vol-ume of 6.5 ml, and the mixture was emulsified by sonication for 1 minute, maintaining the temperature of ; 15 the solution at or below room temperatllre. Where the liposomes were prepared to contain encapsulated [125I]
-;ty~aminyl-inulin, su~h was included in the-phosphate -ibuffer ~ a concentration of about 4 ~Ci/ml buffer.
. . The ether solvent was removed under reduced ~ pressure at room temperature, and the resulting gel was taken up in 1 ml of the above buffer, and shaken vigor-ously. The resulting REV suspension had partiele sizes, as determined by microscopic examination, of between about 0.1 to 20 microns, and was composed predominantly of relatively large (greater than 1 micron) vesicles having one or only a few bilayer lamellae.
The liposomes were extruded twice through a polycarbonate filter (Szoka, 1978), having a selected pore size of 0.4 microns or 0.2 microns. Liposomes extruded through the 0.4 micron filter averaged 0.17 (0.05) micron diameters, and through the 0.2 micron fil-` ter, 0.16 (0.05) micron diameters. Non-encapsulated ~125I] tyraminyl-inulin was removed by passin~ the .
extruded liposomes through Sephadex G-5~ (Pharmacia).
This is a 50,000 molecular weight cut-off gel-extrusion chromatography column.
(*) Trademark "

... . .

.
:. . , ~ :.... ;. , , - ~ ) - 1 32q76~

B. MLVs A total of 10-lOO mg of the selected lipid components were dissolved in chloroform: methanol (2:1).
5 The dissolved lipid was roto-evaporated to a thin film, then hydrated with an aqueous physiological buffer con-taining the solute, e.g., desferal or bleomycin, to be encapsulated. MLVs formed on gentle shaking for 1-2 ; hours. Non-encapsulated solute was removed by gel fil-tration. Examination of the MLVs showed heterogeneous sizes between about 0.1 to 20 microns, and a predomi-nance of multilayered structures. The MLV's were sized by extrusion through a double polycarbonate membrane having a selected pore sizeO The membrane pore size was typically 0.05 micron for low phase transition (fluid) liposome formulations, and 0.08 micron for high phase transition (rigid) liposome formulations. In both ~ cases,-liposome ~izes, as measured by laiser light scat-terin~, were predominantly in the 0.07-0.12 micron size range. When high phase transition lipids were used, rehydration and extrusion were carried at at 50a-60 C.

C. SUVs About 25 ml of the unsized MLV suspension from above was sonicated by untrasonic irradiation using a 1.77cm (1/2 inch) sapphire-bonded probe, with pulse sonication during 15 minute intervals, under optimal output condi-tions. Sonication was carried out under a stream of ni~rogen with the liposome vessel immersed in ice water.
30 The sonicated vesicles were passed through Sephadex G-50 to remove released, free marker. Liposome sizes were predominantly in the 0.03-0.08 micron size range.

;

, ....

:, ., ,. - i : ~ , ,~, ,: : .
, ; '' : , :,~.
, , .
-29- 1 3 2 q 76 9 Example 2 Measurinq Blood/RES Levels Age- and sex-matched mice, typically 20-25 9, were given intravenous injections of liposome suspen-sions or saline by injection into the tail vein. Thetotal amount of material injected was about 0.5 mg phospholipid in a total injection volume of 0.2 ml. At selected time intervals following injection, the animals were sacrificed by cervical dislocation, blood samples were taken from the heart, and the liver and spleen were removed. The liver and spleen were blot dried, weighed and separately counted directly by gamma scintillation counting. A correction factor was applied to account for blood remaining in the liver and spleen. An aliquot of the blood sample was similarly counted by direct gamma counting. Total blood counts were calculated on the basis of a total ~lood volume of 7% body weight.
The ~loodi/RES ratio was calculated as total bloodi countsJto~tal counts of the liver and spleen, and was frequently determined for sevleral other tissues.
~ he blood/RES ratios measured over time were corrected for loss of liposomal radiolabel by multiply-ing the measured blood/RES ratio by the percent of the total counts remaining in vivo at each time point with ' 25 respect to liposomal counts measured immediately after injection.

Example 3 Relationship Between Lipid ComPosition and BloodJRES
REVs sized to 0.17 microns and having the liposome composition and mole ratios indicated at the ~eft ini~a~le l were pi~epared-as in Example-l. The SM
used for the reported studies was bovine brain sphingomyelin, whose hydrocarbon-chain moieties include -- . : .
'': , ' ' ,: ' -30- 1 3 2 q7 6q a mixture of partially unsaturated chains. The liposomes were injected intravenously, and ~he injected animals were sacrificed two h~urs after injection. The table shows liposome sizes, blood/RES ratios, calculated as above, and percent of total of the total encapsulated marker (inulin) recovered at 2 hours post injection.

0 Table 1 Size % remaining ComDosition (~m) Blood/RES in vivo 1 PC 0.17 0.010 + 0.005 86.0 + 5.4 2 PC:CH,2:1 0.17 0.13 ~ 0.08 78.1 t o.o4 3 PC:GMl,1:0.07 0.17 0.17 ~ 0.12 79.4 + 5.9 4 PC:CM:GMl,2:1:0.14 0.16 1.7 + 0.5 75.6 ~ 5.7 PC:CH:ASGMl,2:1:0.14 0.16 0.62 + 0.44 64.8 ~ 1.6 6 DSPC 0.17 0.015 ~ 0.002 91.2 ~ 2.00 7 DSPC:CH,2:1 0.17 0.007 + 0.00 101.2 + 2.4 8 DSPC:GMl,1:0.07 0.17 2.0 + 0.02 76.7 + 3.1 9 DSPC:CH:GMl,2:1:0.14 0.17 3.2 ~ 1.0 64.6 ~ 3.5 lo SM 0.17 0.02 ~ 0.01 27.1 + 3.1 11 SM:CH,2:1 0.17 0.7 ~ 0.2 71.9 + 4.4 12 SM:GMl,1:0.07 0.17 5.7 ~ 1.8 12.4 + 0.7 13 SM:CH:GMl,2:1:0.14 0.17 4.6 ~ 0.6 72.1 ~ 1.5 ~ 14 S~:PC,4:1 0.17 0.6 ~ 0.2 69.0 + 3.3 ? 15 SM:PC:CH,4:1:3 0.17 0.12 ~ 0.06 69.9 ~ 2.2 16 SM:PC:CH:SULF,4:1:3:0.35 0.17 0.43 ~ 0.21 78.4 ~ 1.4 17 SM:PC:GMl,4:1:0.35 0.16 3.3 ~ 0.3 61.8 l 2.9 18 SM:PC:CH:GMl,4:1:3:0.35 0.16 1.5 + 0.6 88.5 ~ 3.0 19 SM:PC:ASGMl,4:1:0.35 0.16 0.9 + 0.5 80.3 ~ 2.5 , 30 The blood/RES ratio data indicated that:
', 1... Ganglioside GM} ~.liposomes also contain-ing:~:a.membrane-rigidifying component, such as .

:
.
` I,.~ .

,:

, ::
cholesterol (composition 4), DSPC (composition 8), and SM (compositions 12, 17), or combinations of these lipid components (compositions 9 and 13) gave much higher blood~RES ratios than liposomes containing ~embrane rigidying components without GMl (compositions 6, 7, lO, 11, 14, or 15), or liposomes containing GMl but without membrane rigidifying components (composition 3).
2. Substituting asilaoganglioside (ASGMl) for GMl in liposomes (composition 19 versus composition 17) largely abolished the GMl enhancement effect on blood/RES ratios.
The percent total recovery data show two SM
formulations (compositions 10 and 12) which have rela-tively poor persistence in the body, presumably because of poor liposome stability and rapid clearance of the .
released encapsulated marker.

Example ~
Effect of DSPC on Blood/RES
Sized REVs having the five lipid compositions , shown at the left in Table 2, were prepared as above.
~, The compositions are similar to several of the Table 1 compositions, except that disteroyl PC (DSPC) has been substituted for the relatively more unsaturated egg PC
(PC)O Blood/RES ratios were determined two hours after IV administration as in Example 3, with the results shown at the right in Table 2. A comparison of ' blood/RES vaues for comparable compositions in Tables 1 and 2 indicates that:
1. Substituting DSPC for PC in an SM:PC:GMl composition .(composit.ion 2 in Table 2 versus -composi-~, tion 17.in~~.Table~ significantly~enhances-the-blood/RES
ratio.- -/

'' ~ .
" ' 1.'~ ;, , 2. Substituting DSPC for PC in the SM:PC:CH:GMl composition (composition 5 in Table 2 ver-sus composîtion lB in Table 1) also enhances the blood/RES ratio.

Table 2 Liposome com~osition Blood/RES ratio DSPC:GMl, 5:0.35 1.8 SM:DSPC:GMl, 4:1:0.35 4.9 SM:DSPC:GMl, 1:1:0.14 3.7 SM:DSPC:GMl, 1:4:0.35 3.1 SM:DSPC:CH:GMl, ~:1:3:0.35 3.7 3xample 5 Time Course of Blood/RES over 24 Hours Blood/RES ratios for two liposome compositions Sized (0.17 micron) REVs containing (1) PC:CH:GMl, 2:1:0.14 (composition 4), and (2j SM:PC~GMl, 4:1:0.35 (composition 17) were e~amined for blood/RES values at 2, 6 and 24 hours post injection.
. The behavior of the two formulations over a 24 hour period is shown in Figure 1 for composition 4 (solid circles) and composition 17 (open circles).
~! 25 As seen, composition 1 was substantially com-' pletely removed after 2 hours, whereas composition 2 retains a significant level in the blood even at 24 , hours post injection.
., Example 6 s Effect of ~anqlioside Concentration on Blood/~ES
Sized MLVs (0.17 microns) containing encapsu-. lated inulin were prepared as above. The liposome com-; positions contained either PC:CH, 2:1 (Figure 2A), or , , ~:2 .

. . .
.
: ' . .

~33- 1 329769 SM:PC, 4:1 (Figure 2B) and increasing molar amounts of GMl, including 0, 2.5, 5.0, 7.5, and 10, 12 and 15 mole percent. The tissue distribution of the liposome label in liver (triangles), spleen (squares), and blood (cir-cles) were determined, as above, two hours post IVadministration.
With reference to to Figure 2~, increasing molar amounts GMl increased blood levels two hours post injection to at most about 10% of total injected counts in liposomes containing 5 and 7.5 mole percent ganglioside, At the same time, the lowest levels of liver uptake which were observed, also at 5 and 7.5 mole percent ganglioside, were greater than 30% of total injected counts.
By contrast, with reference to Figure 2B, increasing amounts of GMl above about 2.5 mole percent increased the percent counts in the blood to above 50%, while total liver uptake fell to less than about 10%.
The iesults demonstrate the importance of membrane-rigidifying components--in this case, SM-- to the effect of GMl on blood/RE5 ratios. The results also show that optimal GMl concentrations of in the liposomes are between about 7.5 and 15.0 mole percent.
:j ExamPle 7 Effect of Suqar and Neqatively Charqed Group~ on - Blood/RES
Sized REVs having one of the 18 different com-positions shown in Table 3 were prepared, to determine ;~ 30 the effect of various sugar and/or negatively charged groups on blood/RES levels 2 hours after liposome admin-istration~ Each of the compositions contained PC:CH, ~:i.and''0;'2 m~Ie'percen~'of one or more'glycolipid or negatively charged lipid. The various charged and~or ~:, ' ~34~ 1 329769 glycolipids used were: monogalactosyl-stearate, sulfatide (SULF) having a charged sulfate group on the galactose residue (Figure 5c): phosphatidylserine (PS);
phosphatidic acid (PA), ceramide trihexosides, (CTRI), an uncharged glycolipid with three hexose units;
digalactosyl diglyceride (DGDG), an uncharged glycolipid with two galactosyl residues; monogalactosyl diglyceride (MGDG), an uncharged glycolipid with a sin-gle galactosyl residue; galactocerebrosides (GAL), an uncharged glycolipid with a single galactosyl residue;
globosides ~GL08), an uncharged glycolipid with 4 sugar residues; asialoganglioside (ASGMl); and glucocerebroside (GLU), an uncharged glycolipid with a single glucose residue.
The molar compositions, and blood/RES ratios measured for 2 hours post injection are given in Table 3 below. In the compositions containing PC and CH, only GMl o~ GAL or GLU in combination with GM1 gave blood/R~S
values which were significantly greater than that given ,20 by PC:CH liposomes. PS and PA both decreased blood/RES
iratios, with PS producing an extreme reduction in blood/RES.
In the compositions containing SM and PC, monogalactosyl-stearate (SULF) gave values comparable to the PC:CH:GMl formulations.

:, : , , ~35~ 1 32976~
.
Table 3 blood/RES, 2 h rs ~ remaining Li~id cOmDOsitiOn~ siZe Dost iniection in vivo (llm~
PC:CH, 2:1 0.1 0.6B~0.15 53.3~6.6 PC:CH:SULF, 2:12:0.2 0.1 0.54+0.22 63.3~1.3 PC:CH:PS, 2:1:0.2 0.1 0.02~0.00 79.2~7.2 PC:CH:PA, 2:1:0.2 0.1 0.09+0.01 62.6~2.2 PC:CH:CTRI, 2:1:0.2 0.1 0.15+0.02 8i.7+2.3 PC:CH:DGDG, 2:1:0.2 0.1 0.74l0.44 43.2~7.2 PC:CH:MGDG, 2:1:0.2 0.1 0.31~0.08 57.4~3.2 PC:CH:GAL, 2:1:0.2 0.1 1.01~0.63 8.7~0.2 PC:CH:GLOB, 2:1:02 0.1 0.08~0.03 7~.2~2.1 PC:CH:ASGMl, 2:1:0.2 0.1 0.46+0.01 75.2~8.1 PC:CH:GLU, 2:1:0.2 0.1 0.72~0.21 84.6~12.1 PC:CH:GMl, 2:1:0.2 0.1 4.3~1.1 75.9~3.1 PC:CH:GAL:GMl, 2:1:0.2 0.1 5.95~1.42 56.8+5.0 PC:CH:GLU:GMl, 2:1:0.2 0.1 3.98+0.91 81.3+3.3 SM:PC, 1:1 0.2 2.14~1.36 72.05~5.57 SM:PC:SULF, 1:1:0.2 0.2 3.0813.41 B2.4+10.8 SM:PC:PA, 1:1:0.2 0.2 1.20+0.31 74.4~2.9 ; SM:PC:PS, 1:1:0.2 0.2 0.02~0.00 81.3t1.8 ~ 20 Blood/R~S values were measured at 2 hours post injection for sized REVs (0.17 micron) composed of SM:PC, 4:1 and 0.35 mole percent of one of the following gangliosides or modified gangliosides: GMl; GM2, con-~, 25 taining one less uncharged terminal sugar residue; GM3, containing two less uncharged sugar residues;
disialoganglioside (GDla~ whose four sugar residues contain two sialic acid moieties and trisialoganglioside (GTlb), whose four sugar residues contain two sialic acid moieties. The blood/RES ratios '3~ are shown in Table 4 below. As seen~ only the GM
~ ganglioside gives high blood/RES ratios.

.~

, - ; . ~ . .............. . .
, : '' .
, :.

1 32976q Table 4 Lipid composition Blood/Res SM:PC:GMl SM:PC:GM2 0.6 (0.3) SM:PC:GM3 0.3 (0~2) SM:PC:GDla 0.6 (0.3) SM: PC: GTlb SM:PC:ASGMl 0.9 (0.5) Example 8 Liposome Distribution 4 Hours Post Iniection MLVs were prepared as in Example 1, using a hydration buffer containing 25 mM desferal. The lipid compositions are listed at the left in Table 5 below, and the relative molar quantities in the adjacent col-umn. The MLVs were extruded as in Example 1, to produce a size range between about 0.07-1.12 microns, and free 20 desferal was removed by SephadexTM G-75 column chromatography.
One day before animal injection, a complex of 67gallium/8-hydroxyquinoline ~a weak lipophilic I chelator) was added to the liposome suspension. When 3 this complex penetrates the liposomes, 67Ga transloca-25 tion to encapsulated desferal occurs. The resulting 7Ga-desferal complex has high affinity and is not dis-placeable by transferrin or other metal-binding pro-teins. If released from liposomes, the complex is rap-idly excreted through the urine with a half life of a 30 few minutes. Immediately before injection, the liposomes are passed through an anion exchange resin (AG-lx4 acetate form) which compleletly removes all non-encapsulated 67Ga-8-hydroxyguinoline complex.

,;, , , ' :' ' : ~ , ~37~ 1 ~2~769 Age- and sex-matched mice were injected intra-venously through the tail vein with a liposome prepara-tion having one of the lipid compositions shown in Table 5 below. Four hours after injection, the levels of radioactivity (counts per minute) in blood and dissected body parts, including liver and spleen were measured by gamma counting, using integral counting between 10 and 1,000 kev. Total blood counts were determined as above, based on an estimated total blood volume. Percent total recovery was determined by whole body gamma counts (whole body counts x 100/injected counts).
The blood/RES ratio was calculated as above, by dividing total blood counts by the sum o total liver and spleen counts, with the results shown in Table 5.
As seen, both GMl ganglioside and hydrogenated PI (HPI) - gave relatively high blood/RES ratios in liposomes con-taining saturated.PC or saturated PC plus SMo The same ganglioside or SULF in an unsaturated liposome formula-tion, or egg PI in an unsaturated formulation gave sub-stantially lower values.

. 25 ~,~
: . 30 .
; ' `- ' .. .

; ,... ..
. .

-38- 1 3~769 Table 5 Liposome Molar Ratio Blood/RES % Total ComDosition of ComPOnents Ratio RecoYery PG:PC:CH 1:10~5 .083 76.1 ~0.8) PG:PC:CH 1:10:5 .006 55.8 (2.9) (unextruded~
GMl:PC:CH 1:10:5 1.1 63.2 ~2.0) SULF:PC:CH 1:10:5 1.1 61.0 (0.9~
DPPG:DSPC:CH 1:10:5 2.0 88.6 (6.1) . PI:PC:CH 1:10:5 .83 49.0 (8.3) . HPI:DSPC:CH 1:10:5 2.5 78.7 (1.5) GMl:SM:DSPC:CH 1:8:2:5 5 50.3 (3.~) GMl:DSPC:CH 1:10:5 5 88.5 (1.2) GL4:PC:CH 1:10:5 .02 --. GTl:PC:CH 1:10:5 .18S --ESxamPle 9 -~ Liposome Distribution 24 hours Post Iniection Sized MLVs having the lipid compositions shown in Table 6 and encapsulating 67gallium/desferal complex were prepared as in ~xample ~. Blood/RES ratios and total percent body recovery, determined as above, are .. shown at the right in the table. The data show that GMl, and HPI in combination with saturated neutral ` lipid components, such as DSPC and SM, give optimal '~! blood/RES ratios at 24 hours.
Lower blood/RES values were obtained where the negatively charged component is PG or DSPG, in which the negative charge is shielded by the relatively small glycerol moiety; and cholesterol sulfate (CHS), in which the charged sulfate group is exposed.

:-.
.

:' .~ ,,j ,, .

1 3~769 Table 6 Liposome Molar Ratio Blood/RES % Total Composition of Components Ratio Recovery GTl:PC:CH 1:10:5 0.004 58.2 (8.8) PG:PC:CH
(unextruded~ 1:10:5 0.00~ 31.3 (1.5) GLOB:PC:CH 1:10:5 .008 34.3 (9.2) PG:PC:CH 1:10:5 .008 ~9.8 (3.9) DSPC:CH 10:5 .014 67.6 (4.2) SULF:PC:CH 1:10:5 .02 41.6 (2.5) SM:PC:CH 8:2:5 .03 21.9 (1.4) .
SM:PC 8:2 .03 6.3 (1.2) HPI:DOPC:CH 1:10:5 .06 46.3 (1.8) CHS:DSPC:CH 1:10:5 .08 55.6 (6.0) GMl:SM:PC 1:8:2 .1 15.1 (0.5) PC:CH 10:5 .11 44.4 (2.9) GMl:SM:PC:CH 1:8-2:5 .12 14.4 (1.9) DPPG:DSPC:CH 1:10:5 .2 59.9 (5.5) PG:DSPC:CH 1:10:5 .2 12.9 (1.4~
PI:PC:CH 1:10:5 .28 37.~ (6.3) ~ SULF:DSPC:CH 1:10:5 .3 57-5 (3 7) :l GMl:PC:CH 1:10:5 .33 40.3 ~1.5~
HPI:DSPC:CH 1:10:5 .43 61.6 (4.0) GMl:SM:DSPC.CH 1:8:2:5 .5 36.9 (5.3) HPI:HPC:CH 1:10:5 .55 51.1 (6.7) GMl-DSPC:CH 1:10:5 .9 66.3 (4.2) :.
Also of note are the varied total body recov-eries after 24 hours with the different formulations.
Although there was no identifiable relationship between percent body recovery to blood or RES values, or their ratio, many of the formulations showed relatively low recoveries where the lipid components had widely varying ;

' ~40- 1 32 9 7 69 phase transition temperatures (Tp). Thus, for example, the GMl formulations con~aining SM (Tp about 30C) and DSPC (Tp about 50~C) gave much lower recovery than the formulation without SM, also as observed at 4 hours post 5 injection. Even more striking was the low recovery seen for the ganglioside or non-ganglioside formulations con-taining SM and egg PC (Tp about 0C), with or without cholesterol.
The total radioactivity levels (liposomal 10 marker) contained in the blood, liver and spleen over a 24 hour post injection period were determined for sev-eral of the liposome compositions, shown at the right in ; Table 7 below. These levels were determined as area under the curve (AUC) for levels measured at 2, 4 and 24 15 hours, and thus are related to total levels of marker present in the tissue (blood, liver or spleen) in the period 24 hours post injection. t ~ As seen from the table, high AUC blood/liver ratios were observed for the formulations containing GM
20 or HPI; and PG or DPPG formulations gave much lower ratios, consistent with the blood/RES ratios in Table 6.

Table 7 Blood/
25Liposome Molar Ratio Blood Liver Liver Composition of Components (AUC) _ AUC Ratio ~ .
PG:PC~CH 1:10:5 0.52 7.34 .07 DPPG:DSPC:CH 1:10:5 3.52 4.27 .82 GMl:PC:CH 1:10:5 2.66 1.68 1.58 GMl:SM:DSPC:CH 1:8:2:5 2.21 1.76 1.25 GM~:DSPC:CH~ 10:5 - - 5-,84 -1.95 3.0 HPI:DSPC:CH 1:10:5 3.82 3.31i 1.15 ,, '' To examine whether a statistically significant correlation exists between liposome blood levels on one hand, and RES levels on the other, a linear regression analysis of the data~ using the least squares method, was performed. Figure 3A shows the inverse (negative) ; correlation between blood levels and RES uptake (r=-0.88, p=0.00002). The values were taken from Table 6.

ExamPle 10 Effect of Charqe on Blood/~ES Ratios Sized MLVs containing increasing molar amounts of HPI or PI and either DSPC or PC plus cholesterol, as indicated in Table 8 below, were prepared as above, and examined for blood/RES ratios and total body recovery 24 or 4 hours post injection. As seen, increasing the molar percentage of HPI from about 6 to 16 produced lit-tle change in the blood/RES ratio, whereas the ratio dropped dramatically between ]6 and 30 percent. A simi-lar drop in ratio was observed for the PI formulations,both 4 and 24 hours post injection. The percent PI or HPI had no major effect on tot~1 body recovery after 4 or 24 hours.

~' :, , .
, ~ ; . _; - ,. . . . .
, .

... . .

Table B
Liposome Molar ~atio ~lood/RES ~ Total Composition of ComDOnents % PI Ratio RecoverY
24 ~ours HPI:DSPC:CH 1:10:5 6.3 .5s 62.2 (6.0) HPI:DSPC:CH 2.5:7.5:5 lb.7 .45 sg.g (3.5) HPI:DSPC:CH 4.5:5.5:5 30.0 .008 4B.3 (l.S) PI:PC:CH 1:10:5 6.3 .27 37.4 (6.3) 0 Pl:PC:CH 4:6:5 26.7 .03 53.4 (15.3) 4 Hours PI:PC:CH 1:10:5 6.3 .83 49.0 (8.3) PI:PC:CH 4:6:5 26.7 .07 72.q t2.3) Exampl ll UPtake of Liposomal Marker into Mouse Tumors . Animals were inoculated with J6456 or Bl6 tumor cell lines. The J6456 line is a T-cell derived lymphoma (Gabizon) that will grow in vitro as a cell suspension, and after intraperitoneal injection, as an ascitic tumor. After IV injection, it will metastasize predominantly into the liver and spleen. The Bl6 mela-noma line is a neuroectoderma.l-derived, solid type of tumor which grows as an adherent tumor in vitro, and ; metastasizes mainly in the lungs.
Tumor cells (106 J6456 or 5 x 105 Bl6 cells) : were inoculated intramuscularly (IM) in the hind leg of syngeneic (C57BLJ6 or ~alb/c) female mice. Between 2-3 weeks after inoculation, when tumors weighed approxi-mately 0.5 to 2 g, mice were injected IV with 1 umol phospholipid of one of the liposome compositions shown in Table 9 below. The molar ratios of components is ~he same as for the same-lipid componen~ formulations. The :.

, ..

.
.. . .. .. .

;;, ~ . -~ .. ~ . , _43_ 1 32~769 .
liposomes were sized MLVs prepared as above with encap-sulated gallium/desferal complex, and having sizes pre-dominantly in the 0.07 to 0.12 micron size range.
Complete animal dissection followed 24 hours S after liposome a~minis~ration. The values shown in the table are based on ~7 gallium counts and corrected for blood content of the tissues. The correction factor for ~ blood content in normal tissues and tumor was determined - by examining the distribution of lllIn-oxine-labeled red blood cells in age- and sex-matched tumor-bearing mice.
Ratios were obtained by dividing the percents of injected dose per gram of respective tissues. Body average represents the average liposome uptake per gram body weight, and was calculat~d by dividing the percent of injected dose recovered in the total body (including tumor) by the weight of the animal.
As seen from ~he data in Table 9, for the J6456-inje~ted mice, a steady increase in tumor uptake (up to ~5 fold~ was observed with liposomes selected for longer in vivo circulation times. Values of between about 4-6% of the injected dose were obtained with the I formulations containing GMl ganglioside or with HPI in combination with saturated phospholipids. These values were obtained after correctiny for blood volume in the tumor, as above. ~hen free 67gallium-desferal complex was injected, the t~mor uptake of the marker was less than 0.1% of the injected dose per gram. The data at , the right in the table show a concomitant decrease in l liver-to-tumor ratios, ind;cating that liposomes accumu-late preferentially in tumors as opposed to non-specific enhancement in all body tissues. Progressively higher ~,~t tumor-to-carcass ratios were also seen with the GMl or HPI formulations (data not shown).

. . .

, ' . . ~ .
... ..

Table 9 Percent of Iniected Dose/G (SD) Ratio: Ratio:
Liposome BodyTumorJ Liver/
Composition Tumor Liver Ava 8cdy Avq Tumor PG:PC:CH 0.2 (0.0) 36.4 (6.3) 2.9 (0.7) 0.1 182.0 PG:PC:CH 0.3 21.4 2.3 0.1 71.3 (unextruded) SULF:PC:CH 0.8 13.6 2.0 0.4 17.0 0 DSPC:CH 2.1 (0.3) 36.5 (7.5)2.8 (0.3) 0.8 17.4 SULF:DSPC:CH 2.1 (0.3) 32.1 (4.5~ 2.1 (0.1) 1.0 15.3 CHS:DSPC:CH 2.5 (0.1) 29.7 (1.4) 2.3 (0.2) 1.1 11.9 HPI:DSPC:CH 4.1 (1.1? 37.8 (0.4) 3.0 (0.2) 1.4 9.2 DPPG:DSPC:CH 4.1 (1.6) 38.3 (0.5) 2.8 (0.1) 1.5 9 3 GM1:DSPC:CH 5.3 (0.9) 31.7 (1.4) 3.3 (0.1) 1.6 6 0 GMl:PC:CH 3.5 (0.6) 20.8 (0.9) 2.4 (0.2) 1.5 5.9 Similar conclusions are-drawn from the tumor .. uptake.da.~a in animals inoculated with the B16, shown in Table lO, although tumor uptake increases as a function of liposome composition are less dramatic.

!

. 25 .. . . .. . . . . .

., . .

, . - , . , , ,~ . .
:, ~ :.
-:

-~5-~able 10 Percent of Iniected Dose/G (S~) Ratio: Ratio:
Liposome Tumor/ Liver/
Composition ~umor Liver BodY ~odY Tumor : GMl:PC:CH 2.5 (0.7) 24.3 (1.3) 2.5 (0.1) 1.0 9.7 SULF:DSPC:CH 3.6 (0.8) 23.5 (1.4) 3.1 (0.3) 1.2 6.5 PG:DSPC:CH 1.5 (0.4) 9.8 (0.6) 1.2 (0.2) 1.3 6.5 DSPC:CH 5.~ 33.2 ~.2 1.3 6.1 0 CHS:DSPC:CH 5.7 (0.5) 21.1 (1.9) 3.5 (0.2) 1.6 3.7 DPPG:DSPC:CH 4.9 (0.3) 17.8 (2.2) 2.9 (0.1) 1.7 ~.6 GMl:DSPC:CH 8.4 (0.3) 37.2 (7.2) 4.4 (0.4) 1.9 4.4 HPI:DSPC:CH 5.3 (0.3) 14.1 (0.3) 2.9 (0.1) l.a 2.7 (1:10:5) HPI:DSPC:CH
(2.5:7.5:5) 5.2 (1.5) 19.7 (2.1) 3.3 (0.3) 1.6 3.8 5 HPI:DSPC:CH
(4.5:,5.5:5) .3 (1) 44.1 (1.9) 2.9 (0.1) 0.1 147.0 , - ~ . ....
- - i - ;Linear regression analysis of the data, using least squares-analysis-, was carried out to determine whether a statistically signiEicant correlation exists between liposome blood levels and tumor uptake. Figure 3B shows a direct (positive) correlation between blood ~ levels and tumor uptake of liposomes in both the B6456 ; (closed triangles~ and B16 (closed circles) animals.
The correlation coefficient for the J6456 animals are r=0.89 and p=0.0005, and for the B16 animals, r=0.91 and p=0.004.

Example 12 - 30 UPtake of Indium-Labeled BleomYcin into Mouse Tumors Sized MLVs composed of GMl:DSPC:CH (1:10:5) and containing encapsulated 111In-bleomycin were pre-pared as above. Bleomycin was labelled with lllIn by adding the label to a suspension of bleomycin liposomes .
;
., . .
~ ., : : ~: ..
: ' .. ' '' ' ~ :.
:; / ~ . . ' :. i ,,, :~. , . , , :

one day before liposome administration, to form a ; high-affinity, lllIn-bleomycin complex encapsulated in the liposomes. Immediately before use the liposomes were passed through an anion-exchange resin, as above, to remove non-encapsulated lllIn.
Mice innoculated with J6456 lymphoma or B16 cells were injected IV with liposomes (1 umole phospholpid/animal) or with an equivalent amount of free In-bleomycin. Tissue distribution of the radiolabel, 24 hours post administration, was determined as above, with the results shown in Tables 11 (J6456-infected ani-mals) and Table 12 (B16-infected animals). As seen, the liposomal form of the drug increased drug quantity 10-30 ; fold over free drug.

Table 11 Percent of Tniected Dose /q Tissue - FREE
TISSUE

; TUMOR 0.7 8.2 (1.5) BLOOD 0.9 2.5 (0.9) LI~EX 0.9 44.8 (0.6) BODY AVG 0.5 4.3 (0.1) , ~ 30 ';
~,. .
- - - - - ., , .: , ,, ., :

~.
'' , :

, ~ , :

Table 12 Percent of In~ected Dose / Tissue FREE Liposomal TISSUE ~ LEO lllIn-BLEo TUMOR 0.3 ~0.0) 9.2 (1.9) BLOOD Ool (0~0) 7~4 (0~8) LIVER 0.4 (0.0) 23.0 (1.1) BODY AVG 0.2 (0,0) 4.0 (0.3) Sized HPI:DSPC~CH (1:10:5) liposomes with encapsulated lllIn-bleomycin were prepared as above and injected IV into mice inoculated with J6456 tumor cells, as above. Animals were sacrificed at 4, 24 and 48 hours and tumor, blood and liver levels of radioactivity determined, with the results shown in Table 13 below.
Blood levels of the label dropped rapidly durir.g the 4-48 hour test period. With ~oth liver and tumor, opti-mal levels were observed at 24 hours, although both 4 and 48 hour levels were relatively high.

Table 13 Percent of Injected Dose / Tissue Tissue 4 hours 24 hours ~8 Hours Blood 47.9 (2.53 8 (1.5) 0.7 (0.1) Liver 20.4 (1.6) 30.6 (1.4) 21.4 (0.8) Tumor 7.8 (1.2) 13.7 (1.1) 10.5 (0.7) . .

-48- 1 32 ~ 7 6 q Example 13 Uptake of Doxorubici _ into_J6~56 Mouse Tumors Sized MLVs having one of the three lipid com-positions shown in Table 14 below were prepared as above. Doxorubicin was included in the hydration buffer, at a concentration of about 5 mg/ml, and free drug was removed from the sized liposomes by gel filtra-tion.
Mice innoculated with J6456 lymphoma cells were injected IV with the MLVs (1 umole phospholpid/animal) or with an equivalent amount of free doxorubicin. Tissue distribution of the drug, 24 hours ; post administration, was determined fluorometrically, with the results shown in Tables 13 below. As seen~
drug levels of the drug, expressed as percent of injected dose/g tumor, were similar to free drug for the two liposome compositions (PG:PC:CH and GMll:PG:PC:CH) ~, which do not show significantly enhanced blood/RES
ratios, whéreas the drug level in tumors was enhanced 3-6 fold with GM1:DSPC:CH liposomes which show optional blood/RES ratios.
:
Table 14 Percent of Iniected Dose (DXR)/ Tumor (SD~
; 25 FREE DXR 0.4 (0.1) PG:PC:CH (DXR) (1:10:5) 0.2 (0.0) ~ GMl:PG:PC:CH (DXR) (1:10:5) 0.2 (0.0) ,.j l GMl:DSPC:CH (DXR~ 10:5~ 1.3 (0.1) ; 30 ;1 While specific methods of preparing and using the liposomes of the invention have been illustrated herein, it will be apparent that a variety of different .~ .

:
" -.,, ~ ,, , .
~ ,. . .

lipid compositions, drug-liposome formulations, and liposome treatment methods can be practiced within the scope of the invention.

S

' :

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

Claims (7)

1. A composition comprising liposomes which contain an entrapped pharmaceutical agent and are characterized by:
(a) liposome sizes substantially between about 0.05 and 0.5 microns, (b) liposomes having substantially homogenous-phase bilayers composed of at least about 50 mole percent of a membrane-rigidifying lipid selected from the group consisting of sphingomyelin and neural phospholipids with predominantly saturated acyl chains, and (c) between about 5-20 mole percent saturated phosphatidylinositol.
2. The composition of claim 1, wherein the liposomes are substantially in the 0.07 to 0.12 micron size range.
3. The composition of claim 1, wherein the membrane-rigidifying lipid is brain sphingomyelin liposomes contain sphingomyelin and the liposomes contain sphingomyelin and phosphatidylcholine, at a mole ratio between 2:1 and 4:1.
4. The composition of claim 1, wherein the membrane-rigidifying lipid is substantially phosphatidylcholine with saturated acyl chains.
5. A method of extending the lifetime of liposome in the bloodstream which comprises preparing the liposomes to contain:
(a) substantially homogenous-phase bilayers containing at least about 50 mole percent of a membrane-rigidifying lipid selected from the group consisting of sphingomyelin and neutral phospholipids with substantially saturated acyl chains, and (b) between about 5-20 mole percent saturated phosphatidylinosito.
6. The method of claim 5, wherein the liposomes contain sphingomyelin and phosphatidylcholine, at a mole ratio between 2:1 and 4:1.
7. The method of claim 5, wherein the membrane-rigidifying lipid is substantially phosphatidylcholine with saturated acyl chains.
CA000555256A 1986-12-24 1987-12-23 Liposomes with enhanced circulation time Expired - Fee Related CA1329769C (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US06/946,415 US4837028A (en) 1986-12-24 1986-12-24 Liposomes with enhanced circulation time
US946,415 1986-12-24
US07/132,136 US4920016A (en) 1986-12-24 1987-12-18 Liposomes with enhanced circulation time
US132,136 1987-12-18

Publications (1)

Publication Number Publication Date
CA1329769C true CA1329769C (en) 1994-05-24

Family

ID=26830135

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000555256A Expired - Fee Related CA1329769C (en) 1986-12-24 1987-12-23 Liposomes with enhanced circulation time

Country Status (7)

Country Link
US (1) US4920016A (en)
EP (1) EP0296212B1 (en)
AU (1) AU603860B2 (en)
CA (1) CA1329769C (en)
DE (1) DE3772385D1 (en)
DK (1) DK472488A (en)
WO (1) WO1988004924A1 (en)

Families Citing this family (449)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5723147A (en) * 1987-02-23 1998-03-03 Depotech Corporation Multivesicular liposomes having a biologically active substance encapsulated therein in the presence of a hydrochloride
US5925375A (en) * 1987-05-22 1999-07-20 The Liposome Company, Inc. Therapeutic use of multilamellar liposomal prostaglandin formulations
US5807572A (en) * 1988-02-18 1998-09-15 Depotech Corporation Multivesicular liposomes having a biologically active substance encapsulated therein in the presence of a hydrochloride
US5169636A (en) * 1988-03-17 1992-12-08 Nippon Fine Chemical Co., Ltd. Liposomes
US5422120A (en) * 1988-05-30 1995-06-06 Depotech Corporation Heterovesicular liposomes
US5230900A (en) * 1988-07-01 1993-07-27 The Biomembrane Institute Antibody-mediated and ligand-mediated targeting of differentiation-inducers to tumor cells
JPH0720857B2 (en) * 1988-08-11 1995-03-08 テルモ株式会社 Liposome and its manufacturing method
GB8824593D0 (en) * 1988-10-20 1988-11-23 Royal Free Hosp School Med Liposomes
US5043164A (en) * 1989-01-17 1991-08-27 The University Of Tennessee Research Corporation Blood-stable, cholesterol-free liposomes
US5356633A (en) * 1989-10-20 1994-10-18 Liposome Technology, Inc. Method of treatment of inflamed tissues
US5843473A (en) * 1989-10-20 1998-12-01 Sequus Pharmaceuticals, Inc. Method of treatment of infected tissues
US5013556A (en) * 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
JP2785981B2 (en) * 1989-11-20 1998-08-13 株式会社資生堂 Emulsion composition
US5053406A (en) * 1989-12-13 1991-10-01 Glycomed, Inc. Compositions and methods for preventing and treating rotaviral infections
US5882678A (en) * 1990-01-12 1999-03-16 The Liposome Co, Inc. Interdigitation-fusion liposomes containing arachidonic acid metabolites
US5162361A (en) * 1990-04-10 1992-11-10 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Method of treating diseases associated with elevated levels of interleukin 1
FR2661331A1 (en) * 1990-04-30 1991-10-31 Oreal COSMETIC OR DERMOPHARMACEUTICAL COMPOSITION CONTAINING VESICLES FORMED BY A MIXTURE OF PHOSPHOLIPIDS / GLYCOLIPIDS.
DE4107152C2 (en) * 1991-03-06 1994-03-24 Gregor Cevc Preparations for non-invasive administration of antidiabetics
DE4107153A1 (en) * 1991-03-06 1992-09-10 Gregor Cevc Compsns. for application of active agents
US5298246A (en) * 1991-01-09 1994-03-29 Kanegafuchi Kagaku Kogyo Kabushiki Kaisha Stable pharmaceutical composition and method for its production
WO1993008298A1 (en) * 1991-10-18 1993-04-29 The Wistar Institute Of Anatomy And Biology Soluble variants of type i membrane proteins, and methods of using them
WO1993020802A1 (en) * 1992-04-09 1993-10-28 Northwestern University Acoustically reflective liposomes and methods to make and use the same
US5858399A (en) * 1992-04-09 1999-01-12 Northwestern University Acoustically reflective liposomes and methods to make and use the same
CA2117769C (en) * 1992-04-10 2003-07-22 Shuji Sato Liposome composition
US5455044A (en) * 1993-05-14 1995-10-03 Depotech Corporation Method for treating neurological disorders
US5534241A (en) * 1993-07-23 1996-07-09 Torchilin; Vladimir P. Amphipathic polychelating compounds and methods of use
US5863556A (en) * 1993-08-20 1999-01-26 Euro-Celtique, S.A. Preparations for the external application of antiseptic agents and/or agents promoting the healing of wounds
US6217849B1 (en) 1993-09-29 2001-04-17 Bracco Research S.A. Liposome suspensions as blood pool imaging contrast agents
JP4076026B2 (en) * 1993-10-25 2008-04-16 トランセイヴ,インコーポレイテッド Liposome defensin
US5468499A (en) * 1993-11-15 1995-11-21 Ohio State University Liposomes containing the salt of phosphoramide mustard and related compounds
CN1099868C (en) * 1993-11-16 2003-01-29 斯卡法玛公司 Vesicles with controlled release of actives
FR2712811B1 (en) * 1993-11-26 1996-01-05 Oreal Method for combating adipositis and compositions which can be used for this purpose.
US5543152A (en) * 1994-06-20 1996-08-06 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5741516A (en) * 1994-06-20 1998-04-21 Inex Pharmaceuticals Corporation Sphingosomes for enhanced drug delivery
US5993850A (en) * 1994-09-13 1999-11-30 Skyepharma Inc. Preparation of multivesicular liposomes for controlled release of encapsulated biologically active substances
US5885613A (en) * 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5820873A (en) * 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
ATE198419T1 (en) * 1994-10-14 2001-01-15 Liposome Co Inc ETHERLIPID LIPOSOMES AND THEIR THERAPEUTIC USE
AU4546996A (en) * 1995-02-24 1996-09-11 Bracco Research S.A. Liposome suspensions as blood pool imaging contrast agents
US6358915B1 (en) * 1995-03-07 2002-03-19 George Washington University Methods for inhibiting metastasis
US5834428A (en) * 1995-04-14 1998-11-10 1149336 Ontario Inc. Glucagon-like peptide-2 and its therapeutic use
US5990077A (en) 1995-04-14 1999-11-23 1149336 Ontario Inc. Glucagon-like peptide-2 and its therapeutic use
US6184201B1 (en) * 1995-04-14 2001-02-06 Nps Allelix Corp. Intestinotrophic glucagon-like peptide-2 analogs
US6673364B1 (en) 1995-06-07 2004-01-06 The University Of British Columbia Liposome having an exchangeable component
US5931809A (en) * 1995-07-14 1999-08-03 Depotech Corporation Epidural administration of therapeutic compounds with sustained rate of release
US5855911A (en) * 1995-08-29 1999-01-05 Board Of Regents, The University Of Texas System Liposomal phosphodiester, phosphorothioate, and P-ethoxy oligonucleotides
US6051600A (en) * 1995-09-12 2000-04-18 Mayhew; Eric Liposomal hydrolysis-promoting hydrophobic taxane derivatives
US6107332A (en) 1995-09-12 2000-08-22 The Liposome Company, Inc. Hydrolysis-promoting hydrophobic taxane derivatives
US6153736A (en) 1995-09-27 2000-11-28 The Liposome Company, Inc. Modified ether glyceroglycolipids
US5834025A (en) * 1995-09-29 1998-11-10 Nanosystems L.L.C. Reduction of intravenously administered nanoparticulate-formulation-induced adverse physiological reactions
USRE39042E1 (en) * 1996-02-16 2006-03-28 The Liposome Company, Inc. Etherlipid-containing multiple lipid liposomes
US6007839A (en) * 1996-02-16 1999-12-28 The Liposome Company, Inc. Preparation of pharmaceutical compositions containing etherlipid-containing multiple lipid liposomes
US5942246A (en) * 1996-02-16 1999-08-24 The Liposome Company, Inc. Etherlipid containing multiple lipid liposomes
US5965159A (en) * 1996-02-16 1999-10-12 The Liposome Company, Inc. Etherlipid-containing multiple lipid liposomes
US6667053B1 (en) 1996-02-16 2003-12-23 Elan Pharmaceuticals, Inc. D and L etherlipid stereoisomers and liposomes
US6197333B1 (en) 1996-03-28 2001-03-06 The Board Of Trustees Of The University Of Illinois Materials and methods for making improved liposome compositions
ES2188929T3 (en) 1996-04-12 2003-07-01 Ontario Inc 1149336 GLUCAGON TYPE 2-PEPTIDAL ANALOGS.
AU5571196A (en) * 1996-04-24 1997-11-12 Sdg, Inc. Improved masking of liposomes from res recognition
US6111081A (en) * 1996-05-31 2000-08-29 Baylor College Of Medicine Lactoferrin variants and uses thereof
US7309692B1 (en) * 1996-07-08 2007-12-18 Board Of Regents, The University Of Texas System Inhibition of chronic myelogenous leukemic cell growth by liposomal-antisense oligodeoxy-nucleotides targeting to GRB2 or CRK1
US6284267B1 (en) 1996-08-14 2001-09-04 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
US7368129B1 (en) 1996-08-14 2008-05-06 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
KR100190826B1 (en) * 1996-08-26 1999-06-01 강재헌 A pharmaceutical composition for inhibiting haemolysis and its preparation method
US5997899A (en) 1996-10-01 1999-12-07 Skyepharma Inc. Method for producing liposomes with increased percent of compound encapsulated
US6977244B2 (en) 1996-10-04 2005-12-20 Board Of Regents, The University Of Texas Systems Inhibition of Bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
DK0964690T3 (en) * 1996-10-15 2003-10-20 Liposome Co Inc Peptide-lipid conjugates, liposomes and liposomal drug administration
US6339069B1 (en) 1996-10-15 2002-01-15 Elan Pharmaceuticalstechnologies, Inc. Peptide-lipid conjugates, liposomes and lipsomal drug delivery
US6037125A (en) * 1996-11-05 2000-03-14 Lexicon Genetics Incorporated Disruption of the mammalian RAD51 protein and disruption of proteins that associate with mammalian RAD51 for hindering cell proliferation and/or viability of proliferating cells
US5827533A (en) * 1997-02-06 1998-10-27 Duke University Liposomes containing active agents aggregated with lipid surfactants
CA2296056A1 (en) * 1997-07-10 1999-01-21 Keith Baker Methods for universally distributing therapeutic agents to the brain
US6217886B1 (en) 1997-07-14 2001-04-17 The Board Of Trustees Of The University Of Illinois Materials and methods for making improved micelle compositions
US20050025819A1 (en) * 1997-07-14 2005-02-03 Hayat Onyuksel Materials and methods for making improved micelle compositions
US20020115609A1 (en) * 1997-07-14 2002-08-22 Hayat Onyuksel Materials and methods for making improved micelle compositions
US6306432B1 (en) 1997-09-08 2001-10-23 Chiron Corporation High and low load formulations of IGF-I in multivesicular liposomes
AU735588B2 (en) 1997-09-18 2001-07-12 Pacira Pharmaceuticals, Inc. Sustained-release liposomal anesthetic compositions
US7285288B1 (en) 1997-10-03 2007-10-23 Board Of Regents, The University Of Texas System Inhibition of Bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
US7704962B1 (en) 1997-10-03 2010-04-27 Board Of Regents, The University Of Texas System Small oligonucleotides with anti-tumor activity
US6734171B1 (en) 1997-10-10 2004-05-11 Inex Pharmaceuticals Corp. Methods for encapsulating nucleic acids in lipid bilayers
ES2384094T3 (en) * 1997-11-14 2012-06-29 Pacira Pharmaceuticals, Inc. Production of multivesicular liposomes
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
EP1190705A4 (en) * 1999-06-24 2009-03-18 Kyowa Hakko Kogyo Kk Method of regulating leakage of drug encapsulated in liposomes
GB9918670D0 (en) 1999-08-06 1999-10-13 Celltech Therapeutics Ltd Biological product
BR0016864A (en) * 1999-12-30 2002-09-24 Judith K Gwathmey Iron chelator supply system, and methods of preparing an iron chelator supply system and treatment of iron overload in a mammal in need of treatment
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
MXPA02008361A (en) * 2000-02-28 2004-05-17 Genesegues Inc Nanocapsule encapsulation system and method.
US8088060B2 (en) 2000-03-15 2012-01-03 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
US8460367B2 (en) * 2000-03-15 2013-06-11 Orbusneich Medical, Inc. Progenitor endothelial cell capturing with a drug eluting implantable medical device
WO2001068158A1 (en) 2000-03-15 2001-09-20 Orbus Medical Technologies Inc. Coating that promotes endothelial cell adherence
US20070055367A1 (en) * 2000-03-15 2007-03-08 Orbus Medical Technologies, Inc. Medical device with coating that promotes endothelial cell adherence and differentiation
US20070141107A1 (en) * 2000-03-15 2007-06-21 Orbusneich Medical, Inc. Progenitor Endothelial Cell Capturing with a Drug Eluting Implantable Medical Device
US9522217B2 (en) 2000-03-15 2016-12-20 Orbusneich Medical, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
US20030229393A1 (en) * 2001-03-15 2003-12-11 Kutryk Michael J. B. Medical device with coating that promotes cell adherence and differentiation
JP2004518692A (en) 2000-12-11 2004-06-24 ザ・リサーチ・ファウンデーション・オヴ・ザ・シティ・ユニヴァーシティ・オヴ・ニューヨーク C-glucosyl ether lipid
US6759058B1 (en) * 2001-04-25 2004-07-06 Western Center For Drug Development College Of Pharmacy Western University Of Health Sciences Enteric-coated proliposomal formulations for poorly water soluble drugs
US8658202B2 (en) 2001-04-25 2014-02-25 Western University Of Health Sciences Coated drug delivery formulations
CA2790034A1 (en) 2001-06-21 2003-01-03 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
NZ566396A (en) 2001-10-09 2009-07-31 Isis Pharmaceuticals Inc Antisense modulation of insulin-like growth factor binding protein 5 expressions
DE60225844T2 (en) * 2001-11-02 2009-04-09 The Governors Of The University Of Alberta, Edmonton MICELLE COMPOSITIONS CONTAIN PEGYLATED PHOSPHOLIPIDS AND A PHOTOSENSIBILIZER
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
EP1460898A4 (en) * 2002-01-03 2006-05-24 Univ Texas Wt1 antisense oligos for the inhibition of breast cancer
US20040038303A1 (en) * 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
US20050222061A1 (en) * 2002-04-18 2005-10-06 Schulte Ralf W Means and methods for the specific inhibition of genes in cells and tissue of the cns and/or eye
US7148342B2 (en) * 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
US20040213837A1 (en) * 2002-08-05 2004-10-28 Sankaram Mantripragada Oil-core compositions for the sustained release of hydrophobic drugs
CN1694959B (en) 2002-09-13 2013-09-18 雷普利瑟公司 Non-sequence complementary antiviral oligonucleotides
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
DK2336318T3 (en) 2002-11-13 2013-07-15 Genzyme Corp ANTISENSE MODULATION OF APOLIPOPROTEIN B EXPRESSION
EP1569695B1 (en) 2002-11-13 2013-05-15 Genzyme Corporation Antisense modulation of apolipoprotein b expression
JP5355842B2 (en) 2002-12-31 2013-11-27 ザイダス ビーエスヴィ ファーマ プライベート リミテッド Non-pegylated long circulating liposomes
US8980310B2 (en) * 2002-12-31 2015-03-17 Bharat Serums and Vaccines, Ltd. Non-pegylated long-circulating liposomes
JP2007524349A (en) 2003-01-16 2007-08-30 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Compositions and methods for siRNA inhibition of ICAM-1
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
WO2005030787A2 (en) * 2003-09-29 2005-04-07 Topigen Pharmaceutique Inc. Oligonucleotide compositions and methods for treating disease including inflammatory conditions
WO2005041870A2 (en) 2003-10-24 2005-05-12 Ader Enterprises, Inc. Composition and method for the treatment of eye disease
US20120027841A1 (en) 2004-01-07 2012-02-02 Biomedical Research Models, Inc. Novel mucosal vaccination approach for herpes simplex virus type-2
EP1713938A2 (en) 2004-02-09 2006-10-25 Thomas Jefferson University DIAGNOSIS AND TREATMENT OF CANCERS WITH MicroRNA LOCATED IN OR NEAR CANCER-ASSOCIATED CHROMOSOMAL FEATURES
CA2563329C (en) 2004-04-30 2016-01-26 Orbus Medical Technologies, Inc. Medical device with coating for capturing genetically-altered cells and methods for using same
WO2005117832A1 (en) * 2004-06-03 2005-12-15 Bracco Research Sa Liposomal assembly for therapeutic and/or diagnostic use
US20060115455A1 (en) * 2004-10-22 2006-06-01 Reed Kenneth C Therapeutic RNAi agents for treating psoriasis
WO2006045202A1 (en) * 2004-10-29 2006-05-04 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
WO2006052767A2 (en) * 2004-11-05 2006-05-18 Inex Pharmaceuticals Corporation Compositions and methods for stabilizing liposomal camptothecin formulations
NZ562946A (en) * 2005-04-29 2011-08-26 Univ Louisville Res Found Cell-surface decoration with active agents
US8287923B2 (en) * 2005-06-10 2012-10-16 Medical College Of Georgia Research Institute, Inc. Compositions and methods for treating immune disorders
US9446017B2 (en) 2005-08-11 2016-09-20 Augusta University Research Institute, Inc. Compositions and methods for treating herpes simplex virus
US8481505B2 (en) 2005-09-12 2013-07-09 The Ohio State University Research Foundation Compositions and methods for the diagnosis and therapy of BCL2-associated cancers
US7700567B2 (en) 2005-09-29 2010-04-20 Supergen, Inc. Oligonucleotide analogues incorporating 5-aza-cytosine therein
KR20080086440A (en) 2005-11-01 2008-09-25 알닐람 파마슈티칼스 인코포레이티드 Rnai inhibition of influenza virus replication
US9149543B2 (en) * 2005-12-15 2015-10-06 The Trustees Of The University Of Pennsylvania Methods and models for rapid, widespread delivery of genetic material to the CNS using non-viral, cationic lipid-mediated vectors
AU2006337093B2 (en) * 2005-12-22 2013-03-14 Opko Pharmaceuticals, Llc. Compositions and methods for regulating complement system
EP1968622B1 (en) 2006-01-05 2014-08-27 The Ohio State University Research Foundation Microrna expression abnormalities in pancreatic endocrine and acinar tumors
ES2545118T3 (en) 2006-01-05 2015-09-08 The Ohio State University Research Foundation MicroRNA based methods and compositions for the diagnosis and treatment of solid cancers
US7943318B2 (en) 2006-01-05 2011-05-17 The Ohio State University Research Foundation Microrna-based methods and compositions for the diagnosis, prognosis and treatment of lung cancer
EP2522749A1 (en) 2006-03-02 2012-11-14 The Ohio State University MicroRNA expression profile associated with pancreatic cancer
ES2446362T3 (en) 2006-03-20 2014-03-07 The Ohio State University Research Foundation Traces of microRNA during human megakaryocytogenesis
US7875288B2 (en) * 2006-03-30 2011-01-25 The Research Foundation Of State University Of New York Method for treating blood coagulation disorders
NZ572308A (en) * 2006-03-30 2011-09-30 Univ New York State Res Found Compositions of less immunogenic and long-circulating protein-lipid complexes
WO2007123691A2 (en) * 2006-03-30 2007-11-01 The Research Foundation Of State University Of New York Method for treating blood coagulation disorders
ES2544861T3 (en) 2006-03-31 2015-09-04 Alnylam Pharmaceuticals Inc. Compositions and methods to inhibit the expression of the Eg5 gene
US7691824B2 (en) 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
US20090130212A1 (en) * 2006-05-15 2009-05-21 Physical Pharmaceutica, Llc Composition and improved method for preparation of small particles
RU2008150373A (en) * 2006-05-19 2010-06-27 Топиджен Фармасьютикалз Инк. (Ca) OLIGONUCLEOTIDES INFLUENCING THE EXPRESSION OF PHOSPHODYESTERASIS
WO2007137156A2 (en) 2006-05-19 2007-11-29 Alnylam Pharmaceuticals, Inc. Rnai modulation of aha and therapeutic uses thereof
US7888498B2 (en) 2006-05-22 2011-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
US8598333B2 (en) 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
US20080152654A1 (en) * 2006-06-12 2008-06-26 Exegenics, Inc., D/B/A Opko Health, Inc. COMPOSITIONS AND METHODS FOR siRNA INHIBITION OF ANGIOGENESIS
EP2436785B1 (en) 2006-07-13 2013-09-11 The Ohio State University Research Foundation MIR-29a for diagnosing poor survival prognosis colon adenocarcinoma.
US7872118B2 (en) * 2006-09-08 2011-01-18 Opko Ophthalmics, Llc siRNA and methods of manufacture
WO2008086807A2 (en) 2007-01-19 2008-07-24 Exiqon A/S Mediated cellular delivery of lna oligonucleotides
US20100329664A1 (en) * 2007-01-23 2010-12-30 Lim Dae-Soon Shutter device for camera
EP2905336A1 (en) 2007-03-29 2015-08-12 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of a gene from the ebola
AR066984A1 (en) 2007-06-15 2009-09-23 Novartis Ag INHIBITION OF THE EXPRESSION OF THE ALFA SUBUNITY OF THE SODIUM EPITELIAL CHANNEL (ENAC) THROUGH ARNI (INTERFERENCE RNA)
CA2692171C (en) 2007-06-22 2019-10-22 Randolph Watnick Methods and uses thereof of prosaposin
US7910722B2 (en) 2007-07-05 2011-03-22 Florida State University Research Foundation RNAi therapeutic for treatment of hepatitis C infection
EP2808398A1 (en) 2007-07-31 2014-12-03 The Ohio State University Research Foundation Methods for reverting methylation by targeting DNMT3A and DNMT3B
US7960336B2 (en) 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
AU2008283997B2 (en) 2007-08-03 2014-04-10 The Ohio State University Research Foundation Ultraconserved regions encoding ncRNAs
WO2009023025A1 (en) * 2007-08-13 2009-02-19 Board Of Trustees Of Southern Illinois University Methods for treatment and prevention of ototoxicity by sirna
CN101836112A (en) 2007-08-22 2010-09-15 俄亥俄州立大学研究基金会 Methods and compositions for inducing deregulation of EPHA7 and ERK phosphorylation in human acute leukemias
AU2008296022B2 (en) 2007-09-06 2014-01-23 The Ohio State University Research Foundation MicroRNA signatures in human ovarian cancer
CN106310293A (en) * 2007-09-27 2017-01-11 免疫疫苗技术有限公司 Use of liposomes in a carrier comprising a continuous hydrophobic phase for delivery of polynucleotides in vivo
US20100209452A1 (en) * 2007-10-03 2010-08-19 Immunovaccine Technologies, Inc Compositions comprising an antigen, an amphipathic compound and a hydrophobic carrier, and uses thereof
WO2009049129A1 (en) 2007-10-11 2009-04-16 The Ohio State University Research Foundation Methods and compositions for the diagnosis and treatment of esphageal adenocarcinomas
EP2617828B1 (en) 2007-12-10 2014-09-24 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor VII gene
AU2009221775B2 (en) 2008-03-05 2015-05-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 and VEGF genes
US8314068B2 (en) 2008-04-14 2012-11-20 University Hospitals Of Cleveland P2X7 inhibition of epithelial cancers and papillomas
CA2724179A1 (en) 2008-05-15 2009-11-19 Topigen Pharmaceuticals Inc. Oligonucleotides for treating inflammation and neoplastic cell proliferation
ES2524699T3 (en) * 2008-06-05 2014-12-11 Immunovaccine Technologies Inc. Compositions comprising liposomes, an antigen, a polynucleotide and a vehicle comprising a continuous phase of a hydrophobic substance
AU2009275387B2 (en) 2008-08-25 2010-07-08 Excaliard Pharmaceuticals, Inc. Antisense oligonucleotides directed against connective tissue growth factor and uses thereof
EP2329044B1 (en) 2008-08-27 2016-05-18 Oncotherapy Science, Inc. Prmt1 for target genes of cancer therapy and diagnosis
EP3208337A1 (en) 2008-09-02 2017-08-23 Alnylam Pharmaceuticals, Inc. Compositions for combined inhibition of mutant egfr and il-6 expression
AU2009298879A1 (en) 2008-09-23 2010-04-08 President And Fellows Of Harvard College SIRT4 and uses thereof
EP3584320A1 (en) 2008-09-25 2019-12-25 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
CN106834291B (en) 2008-10-20 2020-09-29 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of transthyretin
WO2010065834A1 (en) 2008-12-04 2010-06-10 Opko Ophthalmics, Llc Compositions and methods for selective inhibition of pro-angiogenic vegf isoforms
AU2009324534B2 (en) 2008-12-10 2015-07-30 Alnylam Pharmaceuticals, Inc. GNAQ targeted dsRNA compositions and methods for inhibiting expression
WO2010084488A1 (en) 2009-01-20 2010-07-29 Ramot At Tel-Aviv University Ltd. Mir-21 promoter driven targeted cancer therapy
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
WO2010105209A1 (en) 2009-03-12 2010-09-16 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
WO2010127271A1 (en) 2009-04-30 2010-11-04 Intezyne Technologies, Incorporated Polymer micelles containing anthracylines for the treatment of cancer
US8524784B2 (en) 2009-04-30 2013-09-03 Intezyne Technologies, Incorporated Polymer micelles containing anthracylines for the treatment of cancer
IT1398857B1 (en) 2009-06-10 2013-03-21 Univ Padova FOSPHOLIPID POLYMERIC CONJUGATES
WO2011020023A2 (en) 2009-08-14 2011-02-17 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
AU2010288810A1 (en) 2009-08-31 2012-03-01 University Of Bremen MicroRNA-based methods and compositions for the diagnosis, prognosis and treatment of tumor involving chromosomal rearrangements
EP2483407A2 (en) 2009-09-30 2012-08-08 President and Fellows of Harvard College Methods for modulation of autophagy through the modulation of autophagy-enhancing gene products
EP4089169A1 (en) 2009-10-12 2022-11-16 Larry J. Smith Methods and compositions for modulating gene expression using oligonucleotide based drugs administered in vivo or in vitro
CN102695723A (en) 2009-10-30 2012-09-26 中枢神经系统治疗学公司 Improved neurturin molecules
ME03091B (en) 2009-12-01 2019-01-20 Translate Bio Inc Delivery of mrna for the augmentation of proteins and enzymes in human genetic diseases
RU2012127772A (en) * 2009-12-04 2014-01-10 ОПКО ОФТЭЛМИКС, ЭлЭлСи COMPOSITIONS AND METHODS FOR INHIBITING VEGF
EP3925670A1 (en) 2009-12-17 2021-12-22 Children's Medical Center, Corp. Saposin-a derived peptides and uses thereof
JP5860029B2 (en) 2010-03-29 2016-02-16 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. SiRNA therapy for transthyretin (TTR) related ocular amyloidosis
WO2011123943A1 (en) 2010-04-09 2011-10-13 Mount Sinai Hospital Methods for treating disorders of the gastrointestinal tract using a glp-1 agonist
CA2801066C (en) 2010-06-02 2021-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
KR20130098301A (en) 2010-07-09 2013-09-04 엑셀리시스, 인코포레이티드 Combinations of kinase inhibitors for the treatment of cancer
WO2012064824A1 (en) 2010-11-09 2012-05-18 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of eg5 and vegf genes
US9072766B2 (en) 2010-11-18 2015-07-07 Beth Israel Deaconess Medical Center, Inc. Methods of treating obesity by inhibiting nicotinamide N-methyl transferase (NNMT)
WO2012075040A2 (en) 2010-11-30 2012-06-07 Shire Human Genetic Therapies, Inc. mRNA FOR USE IN TREATMENT OF HUMAN GENETIC DISEASES
CN103328038A (en) 2010-12-01 2013-09-25 史拜诺莫度雷森公司 Directed delivery of agents to neural anatomy
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
WO2012106508A1 (en) 2011-02-02 2012-08-09 Pfizer Inc. Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
US20140134728A1 (en) 2011-06-01 2014-05-15 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for adjusting expression of mitochondrial genome by microrna
KR101970634B1 (en) 2011-06-02 2019-04-19 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Methods and uses for ex vivo tissue culture systems
EP3354644A1 (en) 2011-06-08 2018-08-01 Translate Bio, Inc. Cleavable lipids
EP2717893B1 (en) 2011-06-08 2019-05-08 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mrna delivery
US9260519B2 (en) 2011-06-17 2016-02-16 President And Fellows Of Harvard College Frizzled 2 as a target for therapeutic antibodies in the treatment of cancer
EP2723865B1 (en) 2011-06-21 2019-03-27 Alnylam Pharmaceuticals, Inc. METHODS FOR DETERMINING ACTIVITY OF RNAi IN A SUBJECT
SG10201800715PA (en) 2011-06-21 2018-02-27 Alnylam Pharmaceuticals Inc Angiopoietin-like 3 (angptl3) irna compostions and methods of use thereof
EP2723351B1 (en) 2011-06-21 2018-02-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein c (proc) genes
MX344807B (en) 2011-06-21 2017-01-09 Alnylam Pharmaceuticals Inc Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes.
WO2012178033A2 (en) 2011-06-23 2012-12-27 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
CA2839451A1 (en) 2011-06-27 2013-01-03 The Jackson Laboratory Methods and compositions for treatment of cancer and autoimmune disease
EP2543370A1 (en) 2011-07-06 2013-01-09 Georgia Health Sciences University Research Institute, Inc. Compositions and Methods for Treating Herpes Simplex Virus
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
EP2739735A2 (en) 2011-08-01 2014-06-11 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
AU2012321022B2 (en) 2011-10-06 2017-03-23 Immunovaccine Technologies Inc. Liposome compositions comprising an adjuvant that activates or increases the activity of TLR2 and uses thereof
CA2852066A1 (en) 2011-10-14 2013-04-18 The Ohio State University Methods and materials related to ovarian cancer
US20140288156A1 (en) 2011-10-27 2014-09-25 Inserm (Institut National De La Sante Et De La Recherche Medicale) Methods for the treatment and diagnosis of atherosclerosis
EP3560509B1 (en) 2011-12-22 2024-01-31 Children's Medical Center Corporation Saposin-a derived peptides and uses thereof
US9737480B2 (en) 2012-02-06 2017-08-22 President And Fellows Of Harvard College ARRDC1-mediated microvesicles (ARMMs) and uses thereof
ES2762873T3 (en) 2012-03-29 2020-05-26 Translate Bio Inc Ionizable Cationic Lipids
EP2831231A1 (en) 2012-03-30 2015-02-04 Isis Pharmaceuticals, Inc. Compositions and methods for modulating tau expression for reducing seizure and modifying a neurodegenerative syndrome
WO2013153082A1 (en) 2012-04-10 2013-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of nonalcoholic steatohepatitis
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
WO2013153139A1 (en) 2012-04-11 2013-10-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and diagnosis of acute leukemia
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
EP2859102A4 (en) 2012-06-08 2016-05-11 Shire Human Genetic Therapies Nuclease resistant polynucleotides and uses thereof
KR102186116B1 (en) 2012-11-20 2020-12-03 스펙트럼 파마슈티컬즈 인크 Improved method for the preparation of liposome encapsulated vincristine for therapeutic use
LT2929031T (en) 2012-12-05 2018-02-12 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
EP3628335B1 (en) 2012-12-07 2023-11-08 Translate Bio, Inc. Lipidic nanoparticles for mrna delivery in the lungs
WO2014107731A1 (en) 2013-01-07 2014-07-10 Biomedical Research Models, Inc. Therapeutic vaccines for treating herpes simplex virus type 2 infections
BR112015022156A2 (en) 2013-03-14 2017-11-14 Isis Pharmaceuticals Inc compositions and methods for modulating tau expression
CN105142676B (en) 2013-03-14 2022-06-28 夏尔人类遗传性治疗公司 CFTR MRNA compositions and related methods and uses
WO2014152966A1 (en) 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Methods for purification of messenger rna
WO2014160129A2 (en) 2013-03-14 2014-10-02 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
EP2970940B1 (en) 2013-03-14 2018-07-25 Translate Bio, Inc. Mrna therapeutic compositions and use to treat diseases and disorders
EP3757570B1 (en) 2013-03-15 2023-10-11 Translate Bio, Inc. Synergistic enhancement of the delivery of nucleic acids via blended formulations
WO2014160871A2 (en) 2013-03-27 2014-10-02 The General Hospital Corporation Methods and agents for treating alzheimer's disease
WO2014182661A2 (en) 2013-05-06 2014-11-13 Alnylam Pharmaceuticals, Inc Dosages and methods for delivering lipid formulated nucleic acid molecules
CN105452463B (en) 2013-05-22 2019-06-21 阿尔尼拉姆医药品有限公司 TMPRSS6 IRNA composition and its application method
BR112015029139B1 (en) 2013-05-22 2022-07-12 Alnylam Pharmaceuticals, Inc DOUBLE-STRAND RNAI AGENT FOR INHIBITING SERPINA1 EXPRESSION IN A CELL, ITS USES, AS WELL AS PHARMACEUTICAL COMPOSITION AND IN VITRO METHOD OF INHIBITING SERPINA1 EXPRESSION IN A CELL
ES2913946T3 (en) 2013-07-08 2022-06-06 The Univ Of Utah Research Foundation Peptide and its use in the treatment of inflammatory disorders
TWI657819B (en) 2013-07-19 2019-05-01 美商Ionis製藥公司 Compositions for modulating tau expression
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
MX2016004230A (en) 2013-10-02 2016-10-21 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the lect2 gene.
SG11201602631XA (en) 2013-10-04 2016-05-30 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the alas1 gene
CN106413811A (en) 2013-10-22 2017-02-15 夏尔人类遗传性治疗公司 Mrna therapy for argininosuccinate synthetase deficiency
MX2016005239A (en) 2013-10-22 2016-08-12 Shire Human Genetic Therapies Mrna therapy for phenylketonuria.
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
AU2014362262B2 (en) 2013-12-12 2021-05-13 Alnylam Pharmaceuticals, Inc. Complement component iRNA compositions and methods of use thereof
WO2015086828A1 (en) 2013-12-12 2015-06-18 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the prevention and treatment of diabetic cardiomyopathy using mir-424/322
CN113057959A (en) 2014-02-11 2021-07-02 阿尔尼拉姆医药品有限公司 Ketohexokinase (KHK) iRNA compositions and methods of use thereof
EA201691696A1 (en) 2014-04-25 2017-03-31 Шир Хьюман Дженетик Терапис, Инк. METHODS OF CLEANING MATRIX RNA
SG10202104570TA (en) 2014-05-22 2021-06-29 Alnylam Pharmaceuticals Inc Angiotensinogen (agt) irna compositions and methods of use thereof
AU2015298571B2 (en) 2014-07-30 2020-09-03 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
SG10201902499VA (en) 2014-09-03 2019-04-29 Genesegues Inc Therapeutic nanoparticles and related compositions, methods and systems
WO2016036918A1 (en) 2014-09-03 2016-03-10 Boehringer Ingelheim International Gmbh Compound targeting il-23a and tnf-alpha and uses thereof
EP3191591A1 (en) 2014-09-12 2017-07-19 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
US9816080B2 (en) 2014-10-31 2017-11-14 President And Fellows Of Harvard College Delivery of CAS9 via ARRDC1-mediated microvesicles (ARMMs)
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CA2968114A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
TW201702218A (en) 2014-12-12 2017-01-16 美國杰克森實驗室 Compositions and methods relating to the treatment of cancer, autoimmune disease, and neurodegenerative disease
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
CA2974369A1 (en) 2015-01-20 2016-07-28 The Children's Medical Center Corporation Anti-net compounds for treating and preventing fibrosis and for facilitating wound healing
AU2016219263B2 (en) 2015-02-13 2022-12-01 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (PNPLA3) iRNA compositions and methods of use thereof
US10487324B2 (en) 2015-02-25 2019-11-26 Washington University Methods to detect motor neuron disease comprising micro-RNAs
US10302629B2 (en) 2015-03-30 2019-05-28 University Of Maryland, Baltimore Compositions and methods for treating cancer by rational targeting of protein translation
WO2016164746A1 (en) 2015-04-08 2016-10-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
KR20180002688A (en) 2015-05-06 2018-01-08 알닐람 파마슈티칼스 인코포레이티드 (F12), calichein B, plasma (Fletcher factor) 1 (KLKB1) and kininogen 1 (KNG1) iRNA compositions and methods for their use
WO2016201301A1 (en) 2015-06-12 2016-12-15 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
EP3310918B1 (en) 2015-06-18 2020-08-05 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
CA2991167A1 (en) 2015-07-02 2017-01-05 Otsuka Pharmaceutical Co., Ltd. Lyophilized pharmaceutical compositions
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
JP2018520683A (en) 2015-07-17 2018-08-02 アルニラム ファーマスーティカルズ インコーポレイテッドAlnylam Pharmaceuticals, Inc. Multi-target single-body conjugates
TWI678213B (en) 2015-07-22 2019-12-01 美商史倍壯製藥公司 A ready-to-use formulation for vincristine sulfate liposome injection
IL256665B2 (en) 2015-07-23 2023-10-01 Boehringer Ingelheim Int Compound targeting il-23a and b-cell activating factor (baff), nucleic acids encoding same and uses of products comprising them
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
WO2017048843A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the alas1 gene
WO2017048620A1 (en) 2015-09-14 2017-03-23 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting patatin-like phospholipase domain containing 3 (pnpla3) and methods of use thereof
IL258821B (en) 2015-10-23 2022-07-01 Harvard College Nucleobase editors and uses thereof
JP2018536689A (en) 2015-12-10 2018-12-13 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Sterol regulatory element binding protein (SREBP) chaperone (SCAP) iRNA compositions and methods of use thereof
US10435682B2 (en) 2016-02-19 2019-10-08 University Of South Florida Arginine deiminase gene therapy for disordered proteins
EP3468985A1 (en) 2016-06-08 2019-04-17 Children's Medical Center Corporation Engineered botulinum neurotoxins
EP3469083A1 (en) 2016-06-10 2019-04-17 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
US11286473B2 (en) 2016-07-08 2022-03-29 Children's Medical Center Corporation Botulinum neurotoxin and its derivatives
IL264565B1 (en) 2016-08-03 2024-03-01 Harvard College Adenosine nucleobase editors and uses thereof
CA3033327A1 (en) 2016-08-09 2018-02-15 President And Fellows Of Harvard College Programmable cas9-recombinase fusion proteins and uses thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11202818B2 (en) 2016-09-14 2021-12-21 President And Fellows Of Harvard College Methods and compositions for modulating erythropoiesis
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
US11730823B2 (en) 2016-10-03 2023-08-22 President And Fellows Of Harvard College Delivery of therapeutic RNAs via ARRDC1-mediated microvesicles
JP2019530464A (en) 2016-10-14 2019-10-24 プレジデント アンド フェローズ オブ ハーバード カレッジ Nucleobase editor AAV delivery
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
GB2605925B (en) 2016-12-23 2023-02-22 Harvard College Gene editing of PCSK9
MA47603A (en) 2017-02-27 2020-01-01 Translate Bio Inc NEW ARNM CFTR WITH OPTIMIZED CODONS
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
EP3592381A1 (en) 2017-03-09 2020-01-15 President and Fellows of Harvard College Cancer vaccine
CN110914310A (en) 2017-03-10 2020-03-24 哈佛大学的校长及成员们 Cytosine to guanine base editor
WO2018176009A1 (en) 2017-03-23 2018-09-27 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
CN110913898B (en) 2017-04-18 2024-04-05 阿尔尼拉姆医药品有限公司 Methods of treating subjects having Hepatitis B Virus (HBV) infection
WO2018204918A1 (en) 2017-05-05 2018-11-08 Ardelyx, Inc. Treatment of hepatic disorders
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
PE20200746A1 (en) 2017-07-06 2020-07-24 Arrowhead Pharmaceuticals Inc IARN AGENTS FOR THE INHIBITION OF THE EXPRESSION OF ALFA-ENAC AND METHODS OF USE
WO2019023680A1 (en) 2017-07-28 2019-01-31 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
MX2020001233A (en) 2017-08-03 2020-07-20 Otsuka Pharma Co Ltd Drug compound and purification methods thereof.
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
JP2021500871A (en) 2017-09-29 2021-01-14 ザ チルドレンズ メディカル センター コーポレーション Neurotoxin-like toxins and their use
WO2019079347A1 (en) 2017-10-16 2019-04-25 The Broad Institute, Inc. Uses of adenosine base editors
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US10538553B2 (en) 2018-01-05 2020-01-21 University Of South Florida Compounds for the treatment of neurodegenerative diseases
US20220307001A1 (en) 2018-02-27 2022-09-29 President And Fellows Of Harvard College Evolved cas9 variants and uses thereof
US10507226B1 (en) 2018-05-02 2019-12-17 University Of South Florida N-amino peptide beta-sheet mimics for the treatment of Alzheimer's disease
EP3797160A1 (en) 2018-05-23 2021-03-31 The Broad Institute Inc. Base editors and uses thereof
WO2020037125A1 (en) 2018-08-16 2020-02-20 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of the lect2 gene
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
KR20210049138A (en) * 2018-08-24 2021-05-04 플래그쉽 파이어니어링 이노베이션스 브이아이, 엘엘씨 Modified plant messenger pack and uses thereof
WO2020051360A1 (en) 2018-09-05 2020-03-12 The Broad Institute, Inc. Base editing for treating hutchinson-gilford progeria syndrome
AU2019344776A1 (en) 2018-09-18 2021-01-21 Alnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA compositions and methods of use thereof
WO2020076173A1 (en) 2018-09-21 2020-04-16 Universidade De Coimbra Nanodelivery tissue targeting at low systemic exposures and methods thereof
US20220389395A1 (en) 2018-10-29 2022-12-08 The Broad Institute, Inc. Nucleobase editors comprising geocas9 and uses thereof
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
US20220282275A1 (en) 2018-11-15 2022-09-08 The Broad Institute, Inc. G-to-t base editors and uses thereof
WO2020113135A1 (en) 2018-11-29 2020-06-04 Flagship Pioneering Innovations V, Inc. Methods of modulating rna
EP3898977A1 (en) 2018-12-20 2021-10-27 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of kcnt1 related disorders
WO2020181202A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to t:a base editing through adenine deamination and oxidation
WO2020181180A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. A:t to c:g base editors and uses thereof
WO2020181195A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through adenine excision
US20220170013A1 (en) 2019-03-06 2022-06-02 The Broad Institute, Inc. T:a to a:t base editing through adenosine methylation
WO2020181178A1 (en) 2019-03-06 2020-09-10 The Broad Institute, Inc. T:a to a:t base editing through thymine alkylation
JP2022523779A (en) 2019-03-12 2022-04-26 プレジデント アンド フェローズ オブ ハーバード カレッジ Methods and compositions for treating cancer
WO2020191239A1 (en) 2019-03-19 2020-09-24 The Broad Institute, Inc. Methods and compositions for editing nucleotide sequences
US20220204975A1 (en) 2019-04-12 2022-06-30 President And Fellows Of Harvard College System for genome editing
EP3956349A1 (en) 2019-04-17 2022-02-23 The Broad Institute, Inc. Adenine base editors with reduced off-target effects
WO2020212586A1 (en) 2019-04-18 2020-10-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment and prognosis of cancer
WO2020236982A1 (en) 2019-05-20 2020-11-26 The Broad Institute, Inc. Aav delivery of nucleobase editors
WO2020252257A1 (en) 2019-06-12 2020-12-17 President And Fellows Of Harvard College Methods and compositions for modulation of an interspecies gut bacterial pathway for levodopa metabolism
US20220315906A1 (en) 2019-08-08 2022-10-06 The Broad Institute, Inc. Base editors with diversified targeting scope
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2021030666A1 (en) 2019-08-15 2021-02-18 The Broad Institute, Inc. Base editing by transglycosylation
WO2021046122A1 (en) 2019-09-03 2021-03-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US20230279373A1 (en) 2019-09-09 2023-09-07 Beam Therapeutics Inc. Novel crispr enzymes, methods, systems and uses thereof
WO2021067747A1 (en) 2019-10-04 2021-04-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021072328A1 (en) 2019-10-10 2021-04-15 The Broad Institute, Inc. Methods and compositions for prime editing rna
EP4051796A1 (en) 2019-11-01 2022-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
CA3155921A1 (en) 2019-11-01 2021-05-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
US20230086199A1 (en) 2019-11-26 2023-03-23 The Broad Institute, Inc. Systems and methods for evaluating cas9-independent off-target editing of nucleic acids
CA3162908A1 (en) 2019-12-03 2021-06-10 Beam Therapeutics Inc. Synthetic guide rna, compositions, methods, and uses thereof
JP2023506181A (en) 2019-12-13 2023-02-15 アルナイラム ファーマシューティカルズ, インコーポレイテッド Human chromosome 9 open reading frame 72 (C9ORF72) iRNA agent compositions and methods of use thereof
CA3166153A1 (en) 2020-01-28 2021-08-05 The Broad Institute, Inc. Base editors, compositions, and methods for modifying the mitochondrial genome
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
EP4100519A2 (en) 2020-02-05 2022-12-14 The Broad Institute, Inc. Adenine base editors and uses thereof
EP4100032A1 (en) 2020-02-05 2022-12-14 The Broad Institute Inc. Gene editing methods for treating spinal muscular atrophy
EP4103714A1 (en) 2020-02-10 2022-12-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
JP2023519274A (en) 2020-03-26 2023-05-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド CORONAVIRUS iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
JP2023520582A (en) 2020-04-06 2023-05-17 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for silencing MYOC expression
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
US20230159933A1 (en) 2020-04-07 2023-05-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing scn9a expression
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
CN115955972A (en) 2020-04-27 2023-04-11 阿尔尼拉姆医药品有限公司 Apolipoprotein E (APOE) iRNA agent compositions and methods of use thereof
EP4143315A1 (en) 2020-04-28 2023-03-08 The Broad Institute Inc. <smallcaps/>? ? ?ush2a? ? ? ? ?targeted base editing of thegene
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE
CA3162416C (en) 2020-05-15 2023-07-04 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231691A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rsi)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150086A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
US20230183707A1 (en) 2020-05-21 2023-06-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
US20230285357A1 (en) 2020-05-27 2023-09-14 The Regents Of The University Of California Compositions and methods for transdifferentiating cells
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
JP2023540429A (en) 2020-07-10 2023-09-25 アンセルム(アンスティチュート・ナシオナル・ドゥ・ラ・サンテ・エ・ドゥ・ラ・ルシェルシュ・メディカル) Methods and compositions for treating epilepsy
WO2022056454A2 (en) 2020-09-14 2022-03-17 President And Fellows Of Harvard College Methods and compositions for treating hpv-positive cancers
AU2021344422A1 (en) 2020-09-16 2023-05-04 Gordon J. Freeman Methods of treating an individual that has failed an anti-pd-1/anti-pd-l1 therapy
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
WO2022076291A1 (en) 2020-10-05 2022-04-14 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
WO2022087041A1 (en) 2020-10-21 2022-04-28 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
EP4237556A2 (en) 2020-10-28 2023-09-06 Children's Medical Center Corporation Truncated modified recombinant adamts13 and uses thereof
EP4256053A1 (en) 2020-12-01 2023-10-11 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
US11278494B1 (en) 2021-01-22 2022-03-22 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
US11033495B1 (en) 2021-01-22 2021-06-15 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
US11357727B1 (en) 2021-01-22 2022-06-14 Pacira Pharmaceuticals, Inc. Manufacturing of bupivacaine multivesicular liposomes
KR20230146048A (en) 2021-02-12 2023-10-18 알닐람 파마슈티칼스 인코포레이티드 Superoxide dismutase 1 (SOD1) IRNA compositions and methods of using them to treat or prevent superoxide dismutase 1- (SOD1-)-related neurodegenerative diseases
JP2024507533A (en) 2021-02-19 2024-02-20 ビーム・セラピューティクス・インコーポレイテッド Recombinant rabies virus for gene therapy
EP4298220A1 (en) 2021-02-25 2024-01-03 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
AU2022245243A1 (en) 2021-03-23 2023-09-28 Beam Therapeutics Inc. Novel crispr enzymes, methods, systems and uses thereof
IL307239A (en) 2021-03-29 2023-11-01 Alnylam Pharmaceuticals Inc Huntingtin (htt) irna agent compositions and methods of use thereof
JP2024514649A (en) 2021-04-16 2024-04-02 ビーム セラピューティクス インク. Genetic modification of hepatocytes
EP4330396A1 (en) 2021-04-29 2024-03-06 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
WO2022256578A2 (en) 2021-06-02 2022-12-08 Beam Therapeutics Inc. Circular guide rnas for crispr/cas editing systems
TW202308663A (en) 2021-06-04 2023-03-01 美商艾拉倫製藥股份有限公司 Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof
WO2022261509A1 (en) 2021-06-11 2022-12-15 The Broad Institute, Inc. Improved cytosine to guanine base editors
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
KR20240037299A (en) 2021-07-23 2024-03-21 빔 테라퓨틱스, 인크. Guide RNA for CRISPR/CAS editing systems
IL309905A (en) 2021-07-23 2024-03-01 Alnylam Pharmaceuticals Inc Beta-catenin (ctnnb1) irna compositions and methods of use thereof
US20230270840A1 (en) 2021-09-08 2023-08-31 Beam Therapeutics Inc. Viral guide rna delivery
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076898A1 (en) 2021-10-25 2023-05-04 The Broad Institute, Inc. Methods and compositions for editing a genome with prime editing and a recombinase
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023081756A1 (en) 2021-11-03 2023-05-11 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Precise genome editing using retrons
WO2023094435A1 (en) 2021-11-23 2023-06-01 Cellectis Sa New tale protein scaffolds with improved on-target/off-target activity ratios
WO2023102537A2 (en) 2021-12-03 2023-06-08 The Broad Institute, Inc. Self-assembling virus-like particles for delivery of nucleic acid programmable fusion proteins and methods of making and using same
WO2023102538A1 (en) 2021-12-03 2023-06-08 The Broad Institute, Inc. Self-assembling virus-like particles for delivery of prime editors and methods of making and using same
WO2023114953A2 (en) 2021-12-17 2023-06-22 Beam Therapeutics Inc. Novel crispr enzymes, methods, systems and uses thereof
WO2023122762A1 (en) 2021-12-22 2023-06-29 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023150181A1 (en) 2022-02-01 2023-08-10 President And Fellows Of Harvard College Methods and compositions for treating cancer
WO2023196772A1 (en) 2022-04-04 2023-10-12 Beam Therapeutics Inc. Novel rna base editing compositions, systems, methods and uses thereof
WO2023196802A1 (en) 2022-04-04 2023-10-12 The Broad Institute, Inc. Cas9 variants having non-canonical pam specificities and uses thereof
WO2023205687A1 (en) 2022-04-20 2023-10-26 The Broad Institute, Inc. Improved prime editing methods and compositions
WO2023212715A1 (en) 2022-04-28 2023-11-02 The Broad Institute, Inc. Aav vectors encoding base editors and uses thereof
WO2023230613A1 (en) 2022-05-27 2023-11-30 The Broad Institute, Inc. Improved mitochondrial base editors and methods for editing mitochondrial dna
WO2023240137A1 (en) 2022-06-08 2023-12-14 The Board Institute, Inc. Evolved cas14a1 variants, compositions, and methods of making and using same in genome editing
WO2023240277A2 (en) 2022-06-10 2023-12-14 Camp4 Therapeutics Corporation Methods of modulating progranulin expression using antisense oligonucleotides targeting regulatory rnas
WO2024040083A1 (en) 2022-08-16 2024-02-22 The Broad Institute, Inc. Evolved cytosine deaminases and methods of editing dna using same
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
WO2024059791A1 (en) 2022-09-16 2024-03-21 Beam Therapeutics Inc. Large serine recombinases, systems and uses thereof
WO2024077267A1 (en) 2022-10-07 2024-04-11 The Broad Institute, Inc. Prime editing methods and compositions for treating triplet repeat disorders

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2870179A (en) * 1954-08-10 1959-01-20 American Lecithin Co Hydrogenation of phosphatides
US4460577A (en) * 1977-09-30 1984-07-17 Farmitalia Carlo Erba S.P.A. Pharmaceutical compositions consisting or consisting essentially of liposomes, and processes for making same
US4598051A (en) * 1980-03-12 1986-07-01 The Regents Of The University Of California Liposome conjugates and diagnostic methods therewith
JPS57146710A (en) * 1981-03-07 1982-09-10 Kunio Yagi Blood brain-barrier permeating ribosome-like microcapsule
US4416872A (en) * 1982-03-17 1983-11-22 The United States Of America As Represented By The Secretary Of The Army Treatment of malaria with liposomes containing 8-aminoquinoline derivatives and glycoconjugates
US4684625A (en) * 1982-07-08 1987-08-04 Syntex (U.S.A.) Inc. Method for enhancing the anti-infective activity of muramyldipeptide derivatives
US4515736A (en) * 1983-05-12 1985-05-07 The Regents Of The University Of California Method for encapsulating materials into liposomes
US4565696A (en) * 1983-08-03 1986-01-21 The Regents Of The University Of California Production of immunogens by antigen conjugation to liposomes
DE3410238A1 (en) * 1984-03-21 1985-10-03 Bayer Ag, 5090 Leverkusen GLYCOLIPID-LIKE SUBSTANCES IN LIPOSOME FORM
DE3778972D1 (en) * 1986-06-12 1992-06-17 Liposome Co Inc MEDIUM USING LIPOSOME-ENCLOSED, NON-TEROIDER, ANTI-INFLAMMATORY MEDICINAL PRODUCTS.
AU609711B2 (en) * 1986-12-23 1991-05-09 Liposome Company, Inc., The Liposome preparation and antibiotic

Also Published As

Publication number Publication date
WO1988004924A1 (en) 1988-07-14
EP0296212B1 (en) 1991-08-21
US4920016A (en) 1990-04-24
DE3772385D1 (en) 1991-09-26
EP0296212A1 (en) 1988-12-28
EP0296212A4 (en) 1989-03-23
AU1107988A (en) 1988-07-27
DK472488D0 (en) 1988-08-24
DK472488A (en) 1988-08-24
AU603860B2 (en) 1990-11-29

Similar Documents

Publication Publication Date Title
CA1329769C (en) Liposomes with enhanced circulation time
US4837028A (en) Liposomes with enhanced circulation time
DE69531701T2 (en) SPHINGOSOME WITH IMPROVED DRUG RELEASE
US5013556A (en) Liposomes with enhanced circulation time
US5225212A (en) Microreservoir liposome composition and method
Gabizon et al. Liposome formulations with prolonged circulation time in blood and enhanced uptake by tumors.
Gabizon et al. The role of surface charge and hydrophilic groups on liposome clearance in vivo
Fidler et al. Design of liposomes to improve delivery of macrophage-augmenting agents to alveolar macrophages
EP0260811B1 (en) Phospholipid particles encapsulating polyene antibiotics for the treatment of systemic fungal infections
JP4555569B2 (en) Lipid carrier composition having enhanced blood stability
EP0152449B1 (en) Lipid vesicles prepared in a monophase
US20040071768A1 (en) Compositions and methods for treating cancer
WO1987004592A1 (en) Controlled-release liposome delivery system
WO1993019738A1 (en) Method of treatment of infected tissues
Storm et al. Tolerability of liposomes in vivo
Steerenberg et al. Liposomes as drug carrier system for cis-diamminedichloroplatinum (II) II. Antitumor activity in vivo, induction of drug resistance, nephrotoxicity and Pt distribution
EP1448165B1 (en) Lipid carrier compositions and methods for improved drug retention
US20030113369A1 (en) Liposomes with enhanced circulation time and method of treatment
US20050008664A1 (en) Compositions and methods related to lipid:emodin formulations
NAGAYASU et al. Effects of fluidity and vesicle size on antitumor activity and myelosuppressive activity of liposomes loaded with daunorubicin
CA2069635C (en) Fat emulsion
US20010051183A1 (en) Liposomes with enhanced circulation time and method of treatment
Gregoriadis Liposomes as Carriers of Drugs Observations on Vesicle Fate after Injection and Its Control
MXPA05007102A (en) Non-pegylated long-circulating liposomes.
June Liposomes with enhanced circulation time

Legal Events

Date Code Title Description
MKLA Lapsed