CA1333872C - Cancer treatment - Google Patents

Cancer treatment

Info

Publication number
CA1333872C
CA1333872C CA000612992A CA612992A CA1333872C CA 1333872 C CA1333872 C CA 1333872C CA 000612992 A CA000612992 A CA 000612992A CA 612992 A CA612992 A CA 612992A CA 1333872 C CA1333872 C CA 1333872C
Authority
CA
Canada
Prior art keywords
blood
catheter
balloons
plastic tube
organ
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 - Lifetime
Application number
CA000612992A
Other languages
French (fr)
Inventor
William L. Bodden
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.)
BGH MEDICAL PRODUCTS
Delcath Systems Inc
Original Assignee
BGH MEDICAL PRODUCTS
Delcath Systems Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by BGH MEDICAL PRODUCTS, Delcath Systems Inc filed Critical BGH MEDICAL PRODUCTS
Application granted granted Critical
Publication of CA1333872C publication Critical patent/CA1333872C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M25/0067Catheters; Hollow probes characterised by the distal end, e.g. tips
    • A61M25/0074Dynamic characteristics of the catheter tip, e.g. openable, closable, expandable or deformable
    • A61M25/0075Valve means
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3615Cleaning blood contaminated by local chemotherapy of a body part temporarily isolated from the blood circuit
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M1/00Suction or pumping devices for medical purposes; Devices for carrying-off, for treatment of, or for carrying-over, body-liquids; Drainage systems
    • A61M1/36Other treatment of blood in a by-pass of the natural circulatory system, e.g. temperature adaptation, irradiation ; Extra-corporeal blood circuits
    • A61M1/3621Extra-corporeal blood circuits
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M25/10Balloon catheters
    • A61M25/1011Multiple balloon catheters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/10Location thereof with respect to the patient's body
    • A61M60/104Extracorporeal pumps, i.e. the blood being pumped outside the patient's body
    • A61M60/109Extracorporeal pumps, i.e. the blood being pumped outside the patient's body incorporated within extracorporeal blood circuits or systems
    • A61M60/113Extracorporeal pumps, i.e. the blood being pumped outside the patient's body incorporated within extracorporeal blood circuits or systems in other functional devices, e.g. dialysers or heart-lung machines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/20Type thereof
    • A61M60/205Non-positive displacement blood pumps
    • A61M60/216Non-positive displacement blood pumps including a rotating member acting on the blood, e.g. impeller
    • A61M60/226Non-positive displacement blood pumps including a rotating member acting on the blood, e.g. impeller the blood flow through the rotating member having mainly radial components
    • A61M60/232Centrifugal pumps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/30Medical purposes thereof other than the enhancement of the cardiac output
    • A61M60/31Medical purposes thereof other than the enhancement of the cardiac output for enhancement of in vivo organ perfusion, e.g. retroperfusion
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/30Medical purposes thereof other than the enhancement of the cardiac output
    • A61M60/36Medical purposes thereof other than the enhancement of the cardiac output for specific blood treatment; for specific therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/30Medical purposes thereof other than the enhancement of the cardiac output
    • A61M60/36Medical purposes thereof other than the enhancement of the cardiac output for specific blood treatment; for specific therapy
    • A61M60/37Haemodialysis, haemofiltration or diafiltration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/50Details relating to control
    • A61M60/508Electronic control means, e.g. for feedback regulation
    • A61M60/515Regulation using real-time patient data
    • A61M60/531Regulation using real-time patient data using blood pressure data, e.g. from blood pressure sensors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/50Details relating to control
    • A61M60/508Electronic control means, e.g. for feedback regulation
    • A61M60/538Regulation using real-time blood pump operational parameter data, e.g. motor current
    • A61M60/554Regulation using real-time blood pump operational parameter data, e.g. motor current of blood pressure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/80Constructional details other than related to driving
    • A61M60/802Constructional details other than related to driving of non-positive displacement blood pumps
    • A61M60/818Bearings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/80Constructional details other than related to driving
    • A61M60/802Constructional details other than related to driving of non-positive displacement blood pumps
    • A61M60/833Occluders for preventing backflow
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M60/00Blood pumps; Devices for mechanical circulatory actuation; Balloon pumps for circulatory assistance
    • A61M60/80Constructional details other than related to driving
    • A61M60/845Constructional details other than related to driving of extracorporeal blood pumps
    • A61M60/849Disposable parts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M2025/0001Catheters; Hollow probes for pressure measurement
    • A61M2025/0003Catheters; Hollow probes for pressure measurement having an additional lumen transmitting fluid pressure to the outside for measurement
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M25/00Catheters; Hollow probes
    • A61M25/10Balloon catheters
    • A61M2025/1043Balloon catheters with special features or adapted for special applications
    • A61M2025/1052Balloon catheters with special features or adapted for special applications for temporarily occluding a vessel for isolating a sector
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/33Controlling, regulating or measuring
    • A61M2205/3331Pressure; Flow
    • A61M2205/3344Measuring or controlling pressure at the body treatment site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61MDEVICES FOR INTRODUCING MEDIA INTO, OR ONTO, THE BODY; DEVICES FOR TRANSDUCING BODY MEDIA OR FOR TAKING MEDIA FROM THE BODY; DEVICES FOR PRODUCING OR ENDING SLEEP OR STUPOR
    • A61M2205/00General characteristics of the apparatus
    • A61M2205/33Controlling, regulating or measuring
    • A61M2205/3331Pressure; Flow
    • A61M2205/3355Controlling downstream pump pressure

Abstract

Perfusing a high concentration of an agent to treat an organ, such as anti-cancer agents through a body organ containing a tumor, without their entering the body's general circulation, removing them from the organ with effluent blood and transporting the contaminated blood to an extracorporeal circuit where the blood is treated to remove the contamination, and returning the treated blood to the body. The process prevents toxic levels of the agents from entering the body's general circulation while delivering lethal doses of the agents to the tumor. There are described various apparatus for effecting the intra- and extracorporeal treatment of such contaminated blood.

Description

BGHOO I
Cancer Treatment Brief Description of The Invention The proce~s of perfusing a high concentration of anti-cancer a~ents through a body organ cont~ining a tumor without cont~min~tin~ the Dody's 5 general circulation, removing them from the organ wi~h ef~luent blood, transporting the cont~min~ted blood to an extracorporeal circuit, treating the blood in the extracorporeal circuit to remove the cont~min~tion~ and returning the treated blood to the body. The process prevents toxic levels of the agents from entering the body's general circulation while delivering lethal doses of 10 them to the tumor. A variety of apparatus for effecting the intra- and extra- corporeal treatment of such cont~min~ted blood are described.

Background To The Invention Primary cancer of the liver (hepatocellular t.lmor, hepatoma) is a disease with a dismal prognosis due to its relentless pro~es~ion despite many 15 therapeutic modalities. Although uncommon in the United States (ap-proximately 14,000 new cases per year), hepatoma is the most prevalent tumor in the most populous countries of the world. It is quite common in sub-Sahara Africa, Southeast Asia, Japan, the Pacific Islands, Greece and Italy. For those patients not surgically resectable, median survival is approximately 8 weeks.
20 In North A~nerica, this m~ligntqncy most commonly arises in elderly patients with alcoholic or postnecrotic cirrhosis. However, in other parts of the world, it is epidemic and often occurs in young patients. This demographic variation is correlated with a high incidence of early childhood infection with hepatitis B
virus in the geog;aphic areas where hepatocellular tumor is most common.

Although the incidence of primary liver cancer is not high in the United States, cancer of the colon is a major health problem, and cancer of the colon reaches the liver in about 50% of the patients. Over 140,000 new cases are diagnosed yearly. Once the desease has spread, therapy is ineffective, with approximately 50% of all patients dying from their rlice~ce within five years of ., BGHOOl diagnosis. In 15% to 20% of the patients, the tumor will have spread to the liver by the time of diagnosis, and in over 50~ of patients, colon cancer will eventually spread to the liver me~astasis even when there is no tl:mor spread elsewher~. Tumor cells reach the liver via the ~:~rtal ~in arld establi~h a 5 blood supply from the hepatic artery, perhaps through the elaboration of tumor angiogenesis factor(s).

The impact of colon cancer on the liver is grim. When liver metas-tases are diagnosed, the median survival time falls to 4-9 months without treatment. While tumors that originate in other organs do not spread to the 10 liver as frequently, their prognosis is also significantly worsened uhen theyreach the liver. Much medical research assumes that effective treatnent of tumors in the liver will extend survival, improve quality of life, and reduce the financial and emotional irnpact of this desease.

It is a widely held view, and currently being acted upon, as noted 15 below, that effectiveness of chemotherapy is improved by intraarterial infu-sion. However, systemic toxicity has limited drug tolerance. Detoxification of blood cont~ining chemotherapeutic agents has not been developed until this invention.

The current treatment modalities for colon tumor metastatic to the 20 liver are unsatisfactory. A solitary metastatic deposit of colon cancer is best handled by surgical resection, which leads to a 1 year survival rate of 80% and a 3-year survival rate of 40%. However, in 95% of the cases, multiple metas-tatic lesions are present. Systemic chemotherapy has little lasting effect on these metastatic lesions. Although certain drugs have shown activity in 25 various studies, when used at higher doses their effects are negated by theirsystemic toxicities. These same drugs may prove to be much more effective if their systemic toxicities can be avoided. A treatment which exposes a tumor to high antineoplastic drug concentrations and removes the drug from the blood before systemic exposure occurs may be an effective therapy for cancer in the 30 liver.

-1333~72 BGHOO~
At present surgical resection offers the only chance of cure of h~patoma. For resection to be possible, at least one hepatic segment must be spared. The uninvolved segment(s) of liver must be free of cirrhosis. Unfor-tunately, the proportion of patients with potentially resectable tumors is 5 small.

Hepatic artery infusion (HAI) of chemotherapy has been widely investigated. Arterial infusion of 5-FU and FUDR increases their effectiveness by delivering the drug directly to liver tumor cells before its dilution by the systemic circulation. This approach is attractive because hepatocellular 10 tumors frequently remains localized to the liver, and, like most chemotherapeutic drugs, 5-FU display a dose-response effect, i.e., increasing ~- the dose can give a proportionately greater increase in ~ffect. Also, certain drugs, including the fluorinated pyrimidines, doxorubicin and others, are metabolized by the liver and excreted through the biliary tract thereby 15 reducing systemic drug toxicity.

Initially, chemotherapy was given via percutaneously placed catheters with the use of external pumps. Response rates obtained with this form of treatment in patients with colon cancer metastatic to the liver was generally superior than those attained when identical drugs were given intravenously, 20 with objective responses seen in 34% to 83% of the patients. More recent studies, employing surgically placed catheters and implanted pumps, have yielded response rates in 50% to 60% (range 20% to 88%) of the patients with colon cancer metastatic to the liver. The fluorinated pyrimidines (5-FU and FUDR) are the drugs most commonb used for prolonged (over 1 to 2 weeks) 25 HAI, while Mitomycin C and other drugs have been given alone or in combina-tion with these drugs as intermittent bolus injections into the hepatic artery.
To date, no randomized comparative studies have demonstrated that HAI
administration of fluorinated pyrimidines is therapeutically superior to systemically ~tlminictered drug Local and systemic toxicities limit the amount 30 of therapy which can be delivered even by the arterial route.

Local toxicities in the gastrointestinal tract h~ e included gastric and duodenal ulceration, gastric bleeding and/or perforation, severe dyspepsia, gastritis and diarrhea. Many of the ?atients ~ ho developed these local toxicities were found to have had a misplaced or dislodged catheter tip. In 5 these cases, the drug was perfusing a large portion of the stomach and duodenum via the gastric arteries. Gastrointestinal toxicities did not occur when the gastric arteries were separated and ligated from the hepatic artery or embolized at the time of catheter pl~celnent. Diarrhea, a systemic toxicity of the fluorinated pyrimidines, occurs more commonly in patients with arterial 10 to venous (A-V) shunting of 30% or greater A-V shunting allow~ drug to bypass functioning liver cells and avoid being metabolized by the liver thereby increasing systemic drug exposure.

Local hepatobiliary toxicities (hepatitis, cholecystitis, biliary sclerosis, stenosis and stricture) occur in up to 50% of the patients treated with conven-15 tional HAI 5-FU or FUDR chemotherapy. The gallbladder and biliary tree receive all of their blood supply from the hepatic artery whereas ~he liver receives approximately one-fourth of its blood supply from the hepatic artery.
Biliary tract toxicity seems to be more common in patients who have had the blood supply to the biliary tree disrupted by ligation of the gastric arteries.
20 Choliangiography, CT sc~nning and ~lk~line phosphatase elevations have been shown to be effective monitoring tools for identifying patients with impending biliary tract toxicity. Hepatitis, manifested by nausea, vomiting, abdominal pain and jaundice in association with elevated serum concentrations of liver tr~n~min~ces and bilirubin has occurred in patients receiving conventional 25 HAI 5-FU or FUDR. Hepatitis appears to be related to the dose and duration of the hepatic arterial drug infusion.

Systemic toxicity of HAI chemotherapy has not been a major problem when drugs with a high liver extraction ratio, such as 5-FU and FUDR, have been given in conventional doses that are defined by systemic toxicity. Drugs 30 which are not substantially metabolized by the liver upon first pass often cause 1. Toxicity resulting from un;~ ti~n~ ection of drug into an arte~y other than a hepatic artery, most often gastric or ~llo~er~nl branches.

- BGHOOl system toxicities, primarily myelosuppression, when given intraarterially.
Obviously, without the use of a detoxif~ing system that removes unmetaboli-zed drug, most drugs cannot be empl(,sed in higher, potentially more effective, doses by the HAI route.

Systemic chemotherapy for hepatocellular tumor remains a therapeutic challenge. Numerous agents have been tested in Phase II trials;
objective responses to therapy are uncommon. 5-FU and doxorubicin (Adriamycin) are the only dru~s which have consistently been shown to have significant activity. Initial reports of East African blacks treated with doxorubicin, 75mg/m every 3 weeks, resulted in 22 patients attflinin~ an objective response (3 complete) in 50 patients treated. Substantial toxicity occurred with the use of doxorubicin at this dose. Hence, most other studies report on the use of doxorubicin, 60mg/m every 3 weeks. At this dose,.
objective therapeutic responses occurred in appro~im~tely 20% of the patients.
.
Allsm~n, R. K (1961) Development of a technic for isolated perfusion of the liver, NY State J. Med., vol. 61, p. 3993. discloses isolating the liver by curgically separating the portion of the inferior vena cava which includes the hepatic veins, infusing a chemotherapy agent to the liver through the splenic and common hepatic arteries, and collecting the chemotherapy agent from the 20 isolated portion of the inferior vena cava. This reference does not disclose a method for detoxifying blood of chemotherapeutic agent.

K Schwemmle and K Aigner, Recent Results in Cancer Research. vol.
100, pp. 229-233, pub. by Springer-Verlag, Berlin, 1986, utilized isolated hepatic perfusion in two patients suffering from disseminated hepatic metas-25 tases of colorectal cancer. They characterize their work, and that carried outprior to their efforts, as follows:

"Among the various treatment modalities for liver metastases such as resection, intraarterial infusion, isolated perfusion, or chemoe~nboli7~-tion, isolated perfusion enables chemotherapeutic agents to be added to 30 the perfusion circuit in dosages higher than could be tolerated by sys-temic ?riministration. The upper limit of dosage is only the local toxicity.
Because hepatic metastases are mainly vascularized by the hepatic artery, intraarterial infusion of anti-cancer agents provides a much BGHOOI
higher concentration of the drug in these tumors than can be achieved by systemic chemotherapy.
"To develop a method for intraarterial treatment with m~im~l doses of chemotherapeutic drugs, we started with isolated perfusion of the liver 5 in P.nim~l experiments according to previously published methods [4-6'.
Optimal surgical techniques and drug toxicity were studied in dogs.
After these studies in ~nim.ql~ had proved that the method wa~ pr~c-ticable and safe, in November 1981 we performed an isolAted h~l)atjc perfusion in two patients suffering from disseminated hepatic met&s-10 tases of colorectal cancer ~1]. After these two patients had survived 5months without complication, another 38 patients were submitted to isolated hyperthermic perfusion of the liver with chemotherapeutic~ [2].
"In the isolated perfusion circuit both the hepatic artery and portal vein are perfused and the hepatic venous return is collected via a single 15 venous line. During isolated perfusion a portocaval shunt is established in which arnmonium ic filtered out of the portal blood (Fig. 1). Recentl~, we have omitted the filtration unit.
"During the operative procedure through an abdominal midline incision the liver, the hepatoduodenal li~ament, and the inferior caval vein are 20 exposed. Tourniquets are placed around the gastroduodenal arterv and portal vein and around the caval vein below and above the renal veins as well as intrapericardially.
"In order to collect the hepatic venous outflow a double-channel catheter is inserted into the caval vein from below the renal veins. This special 25 catheter consists of a longer channel shunting the caval vein to maintain cardiac venous return and a second shorter channel for the isolated hepatic venous return. The portocaval shunt tube is inserted into the caval vein channel, whereas two lateral openings collect the venous return from the kidneys.
30 "After the perfusion catheter is inserted into the caval vein the portal vein is cannulated in both directions. The peripheral catheter is con-nected to the portocaval filtration unit consisting of a roller pump and a hemofiltrating system. At flow rates of approximately 300-400 ml portal venous blood has been filtered and with adequate volume substitution 35 returned to the caval shunt tube. Thus blood levels of ammoniurn have been kept within normal ranges during the period of isolsted hepatic perfusion. In case of leakage to the systemic circulation a part o~ the anti-cancer drugs as well has been filtered out in that portacaval shunt.
As soon as the shunt is established, the common ~epatic artery is 40 clamped and perfusion is started via the central portal vem catheter in a partial circuit. Then the arterial catheter is inserted into the gastroduodenal artery and the liver is perfused via two arterial lines at a flow rate of 200-350 ml/min in the hepatic artery and 150 ml/min in the portal vein. Heating the perfusion circuit the temperature of the hepatic 45 tissue is increased. The temperature, which should not exceed 40C, is measured with needle probes in the right and left liver lobes. In our first 31 isolated liver perfusions we only applied 5-fluorouracil (5-FU) in a 13338~2 BGHOO~
dosage between 600 and 1000 mg. In the last ei~..t patients a combina-tior. consisting of mito!nycin C ~;nd 5-FU was used.
"Few com lications occurred after iso~ted liver perfusion. Three pa-tients die~ a short tim~ after the operation. One patient died 2 hours 5 later from untreatable bleedin~ a~ter an isols~ed hepatic perfusion combined with a hemihepatectomy. Th~ tumor had already infiltrated the caval vein. In another patient sep-icemia and respir~.tory distr~ss occurred 6 davs after the ~erfusion. The third patient died from ren~l failure 2 weeks after the perfusion. His autopsv showed a 90% regres-10 sion of the tumor Seventy percent of the hepatic tissue in thiC case hadbeerl involved in rnetastases. In the last 30 patients there were no fatal outcomes.
"In spite of the increased survival time unfortunately in the combined group the patients developed extrahepatic metastases after the treat-15 ment. In ten cases (83%) metastases of the lung developed. Half of thepatients developed peritoneal carcinosis or at least positive Iymph nodes at the hepatoduodenal ligament and in three cases (25%) recurrences at the colorectal anastomoses or at the pcrineal scar developed."
"In conclusion we see the following advantages of liver perfusion:
20 1. We are able to use a high anti-cancer dosage which cannot be achieved by other methods, for example, intermittent or continuous intraarterial infusion.
2. The ~flministration of the anti-cancer agents is performed via both the hepatic arteIy and the portal vein.
25 3. A combination with hyperthermia is given.
~The most important disadvantage of this method is the fact that the perfusion cannot be repeated. However, it is possible at any time to continue the therapy with intraarterial infusions and/or with chemoembolization."
(Emphasis supplied) The following publications relate generally to perfusing individual organs with chemotherapy agents: Creech et al. , Healy, Healy, et al.

2. Annals of Surr~en, ~rol. 148, DO. 4, pp. 616-632 (October 1958), note summary at page 632:
'Ch~rnntl~Prapy of canoer has not been entirely ~ fr tory bccause the a~3mir~ie~ration of doses large enough to ci~nifirnntly affect a tumor protuce serious to~ic ef~ects on the bone marrow and ga_trointest~nal tract.~
3. Sur~en-~ynecolo~r Obstetrics, vol. 120, no. 6, pp. 1187-1193 (June, 1965) 4. ~,, vol. I, no. 2, pp. 111-116 (July, 1961) (deals sl,e :r,~ y with liver i~

133387~

Pierpont et al. , and Shingleton, et al. None discloses detoxi~ying blood which has passed through an isolated organ and returning the detoxified blood to the patient.

The following publications disclose applying a chemotherapy agent t~
5 a specific organ, collecting blood generally from the patient, detoxifying theblood, and returning the blood to the patient: Kamidono, et al., and Agishi Krementz, Cancer. vol. 57, no. 3, pp. 416 - 432 (1986), reviewed the development of regional chemotherapy by perfusion. He stated the following re~arding liver perfusion performed surgically in an ~tnim~tl:

10 "Techniques for perfusion of the liver have been complicated, and re-present major abdominal ~-lr~y. Our techniques, developed for the experimental ztnim~l and applicable to patients, mvolved isolation of the liver by p~t~-cin~ a Foley balloon catheter (Bard Urological Division, Murray Hill, NJ) with a ligated tip through the vena cava from the 15 femoral vein to a point proximal to the hepatic veins. The vena cava was occluded above by the balloon that was positioned above the diaphra~rn and below the hepatic veins by a snare placed above the renal veins (Fig.
7), and the hepatic vein drainage was returned to the pump reservoir through the catheter. The hepatic artery was temporarily clamped, and 20 oxygenated blood, and oxygenated blood from the pump and the chemotherapeutic agents were delivered to the liver through the pro-ximal portal vein. Blood from the distal portal vein and vena cava was returned to the heart through an stccessory bypass from the femoral vein to the external jugular vein. We did not persist in our efforts to use 25 hepatic perfusion clinically, but other investigators have separatelY
developed techniques for perfusion of human livers. The approach developed by Ai~ner and colleagues uses a double-lumen tube to collect hepatic venous blood i~ the outer tube, with bypass of the distal caval blood through the i~er tube. Arterial blood and chemotherapy are 5. J. Thoracic and Cardiovas. Sur~.. vol. 39, no. 2. pp. 159-165 (February, 1960) 6. Annals of Sureen, vol. 152, no. 4, pp. 583-593 (October, 1960) 7. The Journal of Urolonr, vol. 131, pp. 3640 (1984) ant Inv~ ;Y9~ UroloYv. vol. 19, No. 3, pp. 176- 178 8. Said to have utilized ~selective delivery of the ~nt j~nr~. drug is achieved by perc~ttsnPo~
injection of 1 mg/kg of mitomycin C into a drug-cbamber of an ~y_.dti._l~ impj9nted vascular access port, another end of which is insertet in the feeding artery of the cancer-bearing organ.~
~Removal of the drug is performed by the usual method of charcoal hc.,.o~y~.ru;~io~ Dr.
Agishi stated. ~However, in order to augment the anticancer erfect, local hyperthermia is fftAbli~l~ed utilizing a radiur~ .,./ wave - 13.56 MHz . emission ..pyar~.tus.~ (Miles Pharm- .'ti~91 Oncology News Update, 1987) 1333872 BGHool delivercd through the hepatic artelg and proximal protal ~sic: portal]
vein to the liver. This method is under study, particularly iri Germany, and has been performed safely with acceptable mor~idit~."
Double balloon catheters in general are described in the following 5 references:

Weikl, et al., U.S. 4,573,966, patented ~iarch 4, 1986, and U.S. 4,610,662, patented September 9, 1986, describe thi.~ use of double balloon catheters to treat stenosis;

Solar, U.S. 4,546,759, patented October 15, 1985, is directed to a triple balloon catheter to assist right ventricle functioning, Hussein, et al., U.S. 4,445,892, patented May 1, 1984, relate to a dual balloon catheter for insertion in blood vessels to provide an isolated operating region in the vessel between the balloons which facilitates the use of an optic system;

Baran, et al., U.S. 4,423,725, patented January 3, 1984, describe a multiple surgical cuff alleged to have a variety of uses.

Betancourt, U.S. 4,180,076, December 25, 1979, describes a nasogastric catheter contS inin~ two infl~tsthle vessels.

German Offenle~ln~schrift 28 34 956 and R~l~si~n Patents 651817 and 511951 describe the use of double balloon catheters for use in isolating the liver for the purpose of blocking blood flow from the liver. The catheters are provided with a bypass to allow blood flow to continue through the ~ 9. Rn~si~n patent 511951 describes the use of a perforated wall catheter for removing blood from the liver and isolating it via a pump, and with respect to the perfusior of the liver with medicants and coolants, the perfusate i9 collected and returned to the liver via a pump.

BGHOOl Implantable pumps have recentl~ ~olne into vogue. However, studies have indicated that a large proportion of the patients developed toxicity due tothe systemic effects of chemotherapy.

In snmm~ry, chemotherapy has not made a dramatic impact on the 5 treatment of primary or metastuti^ liver cancer. Certain drugs and biologicalshave shown considerable activity in various studies, but their effects are negated by systemic toxicity. Some of these may prove to be much more ef~ective if their systemic toxicity can be eliminated.

A treatment which exposes tumors to high concentrations of antineop-10 lastic drugs and biologicals and removes them from the blood before systemicexposure would be an advancein therapy for cancer in the liver. Moreover, it would be desirable to have a method which allows the opportunity for explor-ing HAI therapy with a variety of drugs and biologicals at dosage levels higher than ever before found tolerable by the body A process which allows the 15 variations in the kind and dosage of chemotherapuetic agents to livers would be a significant advance in the treatment of such cancers. A process that does not require general anesthesia or surgery, an.l is suf~lciently non-invasive to allow frequent repetition of therapy would be a significant advance in the art.
There is described herein a process which provides such advantages.
.
The Invention This invention relates to a process of perfusing a high concentration of anti-cancer agents through a body organ cont~ining a tumor without con-t~min~ting the body's general circulation, removing them from the organ with effluent blood, transporting the cont~rnin~ted blood to an extracorporeal 25 circuit, treating the blood in the extracorporeal circuit to remove the con-t~min~tion, and returning the treated blood to the body. The process prevents toxic levels of the agents from entering the body's general circ~ tion while delivering lethal doses of them to the tumor. A variety of apparatus for ef~ecting the intra- and extracorporeal treatment of such cont~min~ted blood 30 are described.

BGNOOI
The process of the invention embraces a system of non-operative an~
sufficiently non-invasive intracorporeal and extracorporeal means to allow frequent repetition of therapy, if desired, which comprises perfusing~lan anti-cancer agent to a tumor, 5 collecting and cont~ining the cont~min~ted blood emanating from the tumor without general circulation of the cont~min~ted blood to the body, transporting the contQ~nin~teA blood from the body to an extracorporeal treatment system, 10 removing anti-cancer agent from the blood in the extracorporeal treat-ment system, and returning the treated blood to the body.

There is described a technique by which anti-cancer agents, such as chemotherapeutic agents, can be removed from the hepatic venous blood 15 before entering the systemic circulation. This permits safe infusion of greater than usual concentrations of anti-cancer agents, such as cytotoxic levels of chemotherapeutic agents, into the hepatic artery for treatment of turnors of the liver. However, the invention in its broadest sense, allows the treatment of a variety of tumor-bearing organs with anti-cancer agents, such as 20 chemotherapeutic agents, while avoiding systemic toxicity.

Tbe invention encornp~ses a process of treating organ site tumors which comprises a. exposing a turnor in a body organ to one or more anti-cancer agents in higher than usual concentrations, 25 b. removing from the organ effluent blood cont~min~ted with the agent provided to the organ, without systemic exposure to the body, BGHOOI
c. passing the efnuent blood from tributa y veins in the organ into a larger vein in which has been provided a catheter cont~ining i. at least one inn~t~sble balloon provided to obstruct passage of the ef-fluent blood to the heart and 5 ii. an avenue, such a plurality of openin~ or a large opening, in the catheter sufficient to accommodate the volume of effluent blood traversing the tribulary veins, d. transporting the cont~smin~ted effluent blood through the catheter and thence from the body into an extracorporeal circuit, 10 e. detoxifying the blood in the extracorporeal circuit, and f. returning the detoxified blood to the body.

More particularly, the invention relates to the treatment of organ site tumors which comprises a. exposing a tumor in a body organ to one or more anti-cancer agents such as 15 antineoplastic drug and biological response modifiers in higher than usual concentrations, b. removing from the organ effluent blood contsmin~sted with the agent provided to the organ, without systemic exposure to the body, c. p~sccing the ef~uent blood from tributa~y veins in the organ into a larger 20 vein in which has been provided a catheter cont~ining i. spaced-apart inflstLs.hle balloons provided to obstruct the large vein above and below said tributary veins and 1 3 3 3 8 7 2 BGHOOl ii. an avenue, such a plurality of openings or a large opening, in the catheter between the balloons sufficient to accommodate the volume of effluent blood traversing the tributaIy veins, d. transporting the cont~min~ted effluent blood through the catheter and 5 thence from the body into an extracorporeal circuit, e. detoxifying the blood in the extracorporeal circuit, and f. returning the detoxified blood to the body.

In respect to the defined treatment, the extracorporeal circuit or - treatment system comprises (a) means for transporting the cont~min~ted 10 blood to (b) means for separating the drug concentration from the blood and also returning the decont~min~ted blood to the body.

The invention has particular importance in the antineoplastic treat-ment of tumors of the liver since anti-cancer levels of antineoplastic agents can be safely infused into the hepatic artery for treatment and systemic toxicity 15 can be avoided. The invention involves intraarterial infusion of the liver with antineoplastic agents and removal of the antineoplastic agents from the hepatic venous blood before it enters the systemic circulation. This invention includes the percutaneous insertion of a special double balloon catheter into the inferior vena cava The catheter contains two inflatable balloons ap~ro~-20 riately spaced to obstruct the inferior vena cava above and below the hepaticveins. Hepatic venous blood is drawn through fenestrations in the catheter wall and thence into an extracorporeal circuit that is openly connected with the catheter's lumen. The blood is decont~min~ted in this circuit and then returned to the systemic circulation via either a subclavian vein, an external 25 jugular veins, the superior vena cava or the right atrium.

The invention includes the use of detoyifi~qtion means, such as one or more of: a hemoperfusion cartridge, hemodialysis, hemofiltration, and hemoadsorbtion through antibodies or biological ligands or molecules able to render them nontoxic and/or to clear the blood of the antineoplastic agent and allow the re-administration of the patient's own detoxified blood.
The invention embraces the passage of the contaminated blood from the double balloon catheter through tubing into a pump that assists the passage of the contaminated blood to a detoxification means such as a hemoperfusion cartridge containing one of a variety of substances, such as a sorbing solid, and/or a hemodialysis unit that removes the drug from the blood. The treated blood is returned to the body via an appropriate large caliber vein.
The invention also includes a disposable kit, that may be used, e.g., for inpatient hospital use with cancer patients, comprising a double balloon catheter, a detoxification means, piping and valves. The kit may include for guidewires, heparin, and other related equipment.
The invention encompasses a double balloon catheter of a. being percutaneously inserted into the inferior vena cava, b. closing off the flow of contaminated blood from the hepatic veins, and c. recovering the contaminated blood from the hepatic veins.
A variety of novel catheters are described that may be used for the recovery of contaminated blood derived from an organ containing a tumor that has been perfused with a anti-cancer agent for its treatment, and for the removal of the contaminated blood from the body so that the blood can be detoxified outside of the body.

133~872 71746-4 In one embodiment, the present invention provides a catheter adapted for percutaneous insertion into a vein or artery, comprising: a plastic tube defining a main lumen for outflowing blood, two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to the cranial end of said plastic tube, and said balloons, when inflated, having a size sufficient to block the flow of blood in a vein or artery into which said catheter is designed to be inserted; fenestrations in said plastic tube between said balloons to said main lumen; and second and third lumina within said plastic tube, said second lumen connecting to the caudal of said balloons and said third lumen connecting to the cranial of said balloons for effecting inflation or deflation of said balloons, the cranial end of said plastic tube being closed to any appreciable inflow of blood.
In another embodiment the present invention provides a kit for removing a treating agent from blood perfused through a body organ of a patient, comprising: a catheter for isolating and removing the blood issuing from said body organ and containing said treating agent; a detoxification means for treating said blood so removed to remove said treating agent; and return catheter means for returning the blood so treated to said patient;
said catheter comprising: a plastic tube defining a main lumen for outflowing blood, two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to the cranial end of said plastic tube and said balloons, when inflated, having a size sufficient to block the . 14a ,~`

~3338~2 71746-4 flow of blood in a vein into which said catheter is designed to be inserted; fenestrations in said plastic tube between said balloons to said lumen and second and third lumina within said plastic tube, said second lumen connecting to the caudal of said balloons and said third lumen connecting to the cranial of said balloons for effecting inflation or deflation of said balloons, the cranial end of said plastic tube being closed to any appreciable inflow of blood.
In yet a further embodiment, the present invention provides a process for removing a treating agent from blood perfused through a body organ of a patient comprising:
isolating blood issuing from said body organ with a catheter;
removing said blood containing said treating agent from said patient through said catheter; detoxifying the blood so removed to remove said treating agent; and returning the blood so detoxified to the patient; said catheter comprising: a plastic tube defining a main lumen for outflowing blood, two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to the cranial end of said plastic tube, and said balloons when inflated having a size sufficient to block the flow of blood in a vein into which said catheter is designed to be inserted; fenestrations in said plastic tube between said balloons to said main lumen and second and third lumina within said plastic tube, said second lumen connecting to the caudal of said balloons and said third lumen connecting to the cranial of said balloons for effecting inflation or deflation of said balloons, the cranial end of said plastic tube being closed 14b 133387~
14c - 71746-4 to any appreclable inflow of blood.
In yet another embodiment the present invention provides an apparatus for perfusing a hlgh concentration of an agent through a body organ requiring treatment without contaminating the body's general circulation, said apparatus comprising - means for removing the agent from the organ with effluent blood;
- means for transporting the contaminated blood to an extracorporeal circuit;
- means for treating the blood in the extracorporeal circuit to remove the contaminatlon; and - meahs to return the treated blood to the body.
In a further embodlment the present invention provides apparatus for isolatlon and treatment of a portion of the body comprising a first catheter adapted for percutaneous insertion into a vein or artery, comprising (a) a plastlc tube havlng a cranlal end and a caudal end, sald plastlc tube defining a maln lumen for outflowing blood; two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to said cranlal end, and sald balloons, when inflated, havlng a slze sufflclent to block the flow of blood in a vein or artery into which said catheter is designed to be lnserted; fenestratlons ln sald plastlc tube between said balloons to sald main lumen; second and third lumlna withln sald plastlc tube, sald second lumen connectlng to one of sald balloons and sald third lumen connecting to the other of said balloons for effecting inflation or deflation of said balloons, said cranial end of said plastic tube being closed to any 14d 13338~ 2 71746-4 appreclable lnflow of blood and (b) a second catheter for returning blood removed through sald maln lumen to the patlent.
Also withln the scope of the present inventlon ls a use of the apparatus of the present invention to treat a body organ without contaminating the body's general circulation.
Brief Description of the Drawlng Flgure 1 shows a diagrammatic and schematic view of signlflcant apparatus features ln relatlonshlp to the body for carrylng out the process of the inventlon.
Flgure 2 shows a diagrammatlc and seml-schematlc vlew of an apparatus assembly for carrylng out the process of the lnventlon.

~:;HOOl rigure 3 shows a partial cross-se^tional side view of one design of a double balloon cathete. useful in the process of the invention.

Figure 4 shows a cross-sectional end view of th~ shaft of the double balloon catheter of Figure 3.

Figure 5 shows a cross-sectional end view of the midsection of a modification of the double balloon catheter of Figure 3.

Figure 6 shows a partial cross-sectional side view of another design of double balloon catheter useful in the process of the invention.

Figure 7 shows a cross-sectional end view of the shaft of the double 10 balloon catheter of Figure 6.

Figure 8 shows a cutaway cross-sectional side view of the interior of a double balloon catheter encompassed by the invention.

Details Of The Invention The process of the invention avoids the use of surgery to isolate the 15 flow of cont~min~ted blood and returns the same~~lood but in a more purified condition to the patient. As a result, the process of the ir:vention may be usedfor extended periods of time, indeed, for periods of time far longer than previously used in the treatment of the same tumors.

The process of the invention is applicable to the treatment of a 20 number of tumors such as those of the kidney, liver, pancreas, bladder and pelvis. Primary and metastatic liver tumors are especially tre~t~ble by the process of this invention. As pointed out above, many of them have shown responsiveness to chemotherapeutic drugs.

In the most preferred case, the invention is directed to the treatment 25 of tumors in the liver by the use of one or more antineoplastic agents, such as chemotherap .etic agents and/or biologicals, and the purification of venous BGHOO~
blood from the liver to avoid systemic circulation of the agent(s). This may involve the use of balloon catheters that are suitable for insertion in the inferior vena cava to isolate venous outflow from the liver and permit the removal of blood cont~min~ted with antineoplastic agent from the body with a 5 pump. The cont~min~ted blood will be filtered through detoxifying means and then returned to the patient via a large caliber systemic vein at a point above the diaphragm. Because primary hepatocellular and metastatic hepatic tumors derive their blood supply from the hepatic artery, the tumor will be perfused by high concentrations of, for example, a chemotherapeutic agent 10 such as 5-FU or biologicals. Because a normal liver receives three-fourths ofits blood supply from the portal vein the drug or biological will be diluted by a factor of about three before it reaches normal, uninvolved liver cells, thereby protecting them against hepatotoxicity.
r As noted above, si~nific~nt elements of this process have been tested 15 in man. In a manner, each component of the system (HAI arterial catheter infusion of chemotherapy, hemodynamic isolation of the hepatic veins by balloon catheters, hemofiltration of ~-FU, and subclavian vein insertion) has been used in h-lm~nc. However, an extracorporeal drug removal approach with hepatic isolation has only been performed by laparotomy.

The Catheters As mentioned above, the ~rocess of the invention involves the per-cutaneous placement of unique double balloon catheter desi~ns. These catheters may use a variety of designs and sizes depending on the organ whose veins are isolated and the sizes and locations of it's veins. The primary 25 function of the double balloon catheter is to isolate the flow of blood from the veins carrying the effluent blood from the organ cont~ining the tumor that is under treatment. Venous isolation precludes systemic perfusion of the cont~min~ted blood. Thus the tip of the double balloon catheter is to be placed in the body so that the venous effluent from the organ being treated is pre-30 vented from flowing to the heart. The space between the two balloons ispredetermined to ensure removing the full quantity of cont~rnin~ted blood from the treated organ. The space between the balloons is large enough that 13~3872 the balloon central in position can be located in a position in the most centraldraining vein to block cont~min~ted venous blood flow to the h~ ~rt and the balloon peripheral in position can be located; ~ripheral in the most central draining vein to block the flow of uncont~min~ed blood to the cont~min~ted 5 venous blood flow. Veins from organs not under treatment can enter the segment between balloons without detrimental efrect as long as the ~ump and filtration system can accommodate the additional volume. The ~ cnous anatomy of the organ under treatment or of adjacent organs can be altered where necessary by obstruction using angiographic embolisation or ablation 10 techniques and materials, including detachable balloons or stainless steel coils.

The lumen of the catheter between the balloons is openly connected, or can be made openly connected, to the surrounding vein In addition,- the same lumen of the catheter is also openly connected, or can be made openly connected, to the extracorporeal circuit, thereby providing free flow of the 15 cont~min~ted blood from the veins to the extracorporeal circuit. Thus the catheter has a main lumen to act as a conduit for the cont~min~ted blood flow from the venous effluent(s) to the extracorporeal circuit.

The size of the main lumen is determined by the material of which it is made, the volume of blood to be transported through it and the diarneter of 20 the vein in which it will be located. The main lumen may be an open annulus or semi-annulus located within the peripheral balloon that is openly colmected to the extracorporeal circuit. In this type of catheter, a central rod or rodlike axis is provided for support for the balloons.

The catheter may also have supplemental l~min~ The supplemental 25 lumina are smaller in size, i.e., in diameter or cross-sectional area, than the main lumen. They may serve any of a number of ancillary functions in the process. For example, in one lesiFn, a supplemental lumen courses through the full length of the catheter for the purpose of accommodating a guidewire that is desirable for percutaneous insertion of the catheter. Each balloon may 30 be provided with a supplemental lumina to be used for its inflation, or one supplemental lumen may be used for supplying nuid for the inflation of both balloons. A~ additional supplemental lumen may be provided for connection BGHOOI
to a pressure monitor to continuously measure the pressure of the venous emuent. This lumen can also be used to inject contrast medium, if provided with a connector that can accommodate an injection device. In some ~1esign~, the main lumen may be used for one or more of the above functions. This 5 multifunctionality can serve to reduce the cost in m~king the catheter and simplify the apparatus. The main and/or supplemental lumina may be made from separate tubing threaded into the catheter or from channels molded into the structure of the catheter. Another supplemental lumen can be used to return detoxified blood to the general circulation and avoid puncture of 10 another vein.

The wall(s) of the segment of the catheter between the balloons is/are provided with fenestrations to allow entry of venous blood into the main lumen. The number, shape and size of the fenestrations may vary according to.
the size of the catheter, the rate and volume of blood they must transmit, and 15 the materials of construction of the catheter. The shape and size of the fenestrations should take into consideration turbulence effects as the blood courses though the fenestrations and into the main lumen. Fenestrations that are too small can elevate hepatic sinusoidal pressure and fenestrations that aretoo large may weaken the catheter walls and compromise the integrity of the 20 catheter.

One practical double balloon catheter design would have one large central lumen, 2 smaller lumina and 2 inflatable balloons that are separated by about 9 to 10 cm. in the length of catheter that contains perforations. The catheter is designed to be positioned (under fluoroscopic guidance) in the 25 inferior vena cava (IVC) such that the central balloon, when inflated, occludes the IVC just above the hepatic veins. The peripheral balloon, when inflated, occludes the IVC just below the hepatic veins, thus isolating hepatic venous blood from the systemic circulation. Perforations in the catheter between the two inflated balloons convey blood through the large central catheter lumen to 30 a variable speed pump and filtering device. An inferior vena cavagram throughthe main lumen can be used to document complete obstruction of the inferior vena cava proarim~l and distal to the hepatic veins. The effectiveness of passage of blood from the liver through the extracorporeal circuit can be BGHOOl monitored by pressure measurement in the central catheter lumen. The variable speed pump is adjusted to maintain normal hepatic vein pressure and flow. The detoxifying means reduce the chemotherapeutic agent such as 5-FU
in the blood to nontoxic levels before the blood is returned to the ~ stemic 5 circulation.

In another design an independent return lumen courses through the main lumen. One end to the return lumen is connected to the outlet of the extracorporeal circuit and the other end openly outlets into a vein at a location superior to the diaphragrn. When the double balloon catheter is located in the 10 IVC, this return lu~nen extends beyond the end of the main catheter to the right atrium. In this construction, the return lumen consists of a separate piece of tubing threaded inside the main lumen and through the end hole of the catheter. The return lumen is large enough to carry the full volume of the.
blood being returned to the patient from the detoxifying apparatus. In 15 another embodiment of the invention, part of the return flow of the effectively detoxified blood is fed through the return lumen and the remainder is separately fed to the patient via a separate feed system, such as through a separate catheter feed to one of the subclav~an veins, as described by Krementz, supra.

The double balloon catheter, once properly located in the body, extends through the skin to the outside of the body. It terrninates in a Luer fitting and a valve cutoff such as a stopcock. The extracorporeal circuit can beseparated from the double balloon catheter and reconnected at will. When the balloons are not inflated, blood flow through the IVC is maintained. When the 25 balloons are inflated, the blood below the peripheral balloon will find secondary pathways to the heart.

This convenience may be duplicated on the supply side of the process, where the chemotherapuetic agent is supplied to the arterial side of the liver, via the hepatic artery, by the percutaneous insertion of a feed catheter to the 30 hepatic artery, leaving the tubular ending of the feed catheter in a plastic reservoir surgically implanted just below the patient's skin and surgically tiedtherein below the skin. The plastic reser~oir contains a resealing membrane BGHOOl of a type simil~r to those used in multi-dose vials that can be percutaneously r)enetrated from the outside of the body by one or more needles to reinitiate the flow of chemotherapuetic agent to the diseased organ. Illustrative of such devices is Implantof~ Drug Delivery System, sold by Burron Medical Inc., 824 5 Twelfth Avenu~ Bethlehem, PA 18018.

The double balloon catheter can be introduced into the femoral vein using the Seldinger technique. A guidewire made of stainle~s steel is first passed through a needle that has been inierted percutaneously into the vein.
A catheter with a single balloon is inserted over the guidewire and the balloon 10 is innated to dilate the percutaneous tract to the diameter of the sheath that will transmit the double balloon catheter. A plastic sheath tubing is passed over the guidewire when the single balloon catheter is removed. After the sheath is properly located in the vein the double balloon catheter is inserted within the sheath and over the guidewire and advanced to the proper position 15 relative to the organ to be treated. All manipulations of the double balloon catheter are done under fluoroscopic control. An inferior vena cavagram can be performed prior to catheter insertion or prior to balloon inflation with the patient lying on an opaque ruler, parallel to the IVC. The hepatic veins and renal veins can be identified and their location determined according to the 20 opaque ruler.
, . . ~
Under fluoroscopic guidance, the catheter is positioned so that the central balloon, when inflated, occludes the IVC just above the hepatic veins.
The peripheral balloon, when inflated, occludes the IVC just below the hepatic veins. Dilute contrast medium such as saline solution is used to inflate the 25 balloons and reference to the ruler insures their accurate positioning.

In a specific embodiment of the invention, the double-balloon catheter contains three lllmin~ One lumen transmits an angiographic guidewire and is used for percutaneous insertion. A main lumen carries hepatic venous blood from the fenestrations between the balloons to the extracorporeal circuit. The 30 third lumen terminates at the fenestrations and is used to measure pressure or inject contrast medium. A pressure monitor, attached to this lumen, measures pressure within the isolated segment of the vena cava before and BGHOol during balloon infl~tion The pres ure measured before balloon inflation is the systemic venous pressure. The pressure measured after balloon inflation but before opening the extracorporeal circuit is equal to the wedge hepatic venous pressure, which is assumed to be equal to portal pressure. This measurement 5 can determine the presence or absence of portal hypertension. The pressure measured after balloon inflation and during flow through the extracorporeal circuit is the hepatic venous pressure. The hepatic venous pressure can be monitored continuously during drug infusion. The speed of the pump in the extracorporeal circuit can be adjusted to maintain hepatic venous pressure 10 above systemic venous pres~lre but below portal pressure. Thi~ prevents hepatic sinusoidal congestion. The caliber of the balloon catheter and of the tubing in the extracorporeal circuit are calculated to ensure that they are of sufficient size to transmit the necessary volumes of blood with minim~l resistance.

After infl~tion of the balloons, an inferior vena cavagrarn (contrast medium injected into the inferior vena cava) is typically performed through the double balloon catheter prior to infusion to document complete obstruction of the vena cava proxirnal and distal to the hepatic veins and to demonstrate the anatomy of the hepatic veins. Samples of hepatic venous blood are 20 generally aspirated through the pressure port of the double balloon catheter immediately after the beginning of infusion, and, in the typical case, at inter-vals not to exceed one hour during infusion, and for at least three hours after infusion, the samples are analyzed for chemotherapy agent concentrations.
Simultaneous blood samples are taken from the extracorporeal circuit after 25 detoxification and analyzed for drug concentrations in order to document the efficiency of the detoxification means in removing the drug from the blood before returning the blood to the systemic circulation. In addition, blood samples are obtained from a peripheral vein to evaluate drug concentrations re~rhing the systernic circulation. Systemic drug concentrations are then 30 measured over 24 to 48 hours following the infusion.

Another double balloon catheter design may utilize only 2 supplemen-tal lumina and one main lumen for blood transfer to the extracorporeal circuit.
Each supplemental lumen can supply fluids to one of the balloons.

13~3~

BGHOO I
Th~ Pump The venous pressure provides the pressure for ~assaFe of b, ~od to ti.e extracorporeal circuit. The function of the pump i~ to continue the movement of blood though the extracorF)oreal circuit and return it to the patient. Th~
5 blood is removed from the body by a combination of gravitational displacement and the venous blood pressure. The pump does not generate a negative pressure and pull blood from the body. The pressure of the return flow of the blood from the extracorporeal circuit to t~.;? systemic veneous system should beless than about 300 mm Hg.

A variety of suitable pumps are comm;~rcially available. They come in a number of designs. A preferred design is a centrifugal cardiopulmonar~r bypass pump that utilizes smooth surface rotators without relying on rotatin~
vanes. These pumps have been used in long term support of cardiac bypass and in liver transplants. Such designs are shown in the following U.S. Patents:

3,487,784, patented Jan. 6, 1970 Reissue 28,742, reissued Mar. 23, 1976 3,647,324, patented Mar. 7, 1972 3,864,055, patented Feb. 4, 1975 3,957,389, patented May 18,1976 3,970,408, patented July 20, 1976 4,037,984, patented July 26, 1977 Such pumps are obt~in~hle from Bio-Medicus, Inc., Minneapolis, MN
55344. This pump reacts to pressure changes automatically, and it has several inherent safety features. The centrifuge is volume dependent and the pump 25 can decrease the flow rate if the venous drainage is interrupted. The pllmp can also slow itself down in instances of too much resistance to flow. For example, at a resistance of 700 mm Hg., the smooth rotator pump reduces its output flow to zero. In the event of power failure, the pump can automatically change over to battery power for uninterrupted pumping. This type of pump 30 does not impose a negative pressure that pulls the blood flowing from the body and does not adversely affect the chemistry of the blood. They are herein - BGHOOl cha.~cterized as smooth rotator pumps and referred to herein and in the claims as a "smooth rotator pump."

Another useful type of pump is the centrifugal pump such as the vsne (impeller) desi~n sol~ by Sarns Inc/:M, Ann Arbor, MI 48106. Another type of 5 pump is the roller bearing pump. The preferred pump for practicing the process of the invention Is the smooth rotator pump.

Blood Detoxification The cont~min~ted blood captured by the double balloon catheter is fed through tubing to the pump and then to a blood detoxification step. The 10 process will be successful even if the anti-cancer agent is not completely removed from the blood. The important point is that the arnount of anti-cancer agent in the body be kept below toxicity levels. One hundred percent removal of any drug is seldom possible and generally not practical. The decont~min~tion of the blood may be effected by a number of standard 15 procedures known to the art. These include the use of detoxification means, such as a hemoperfusion cartridge and/or hemodialysis and/or hemofiltration and/or hemoadsorbtion through antibodies or biological ligands or molecules able to render them nonto-x-ic~ to clear the blood of the antineoplastic agent and allow the re-arlminictration of the patient's own detoxifled blood. The detoxi-20 fication step comprises any process by which the concentration of the anti-cancer agent in the blood can be removed so that the blood can be returned to the body without causing systemic to-x-icity.

Hemoperfusion involves the passage of the cont~min~ted blood over a solid surface detoxicant particulate mass that separates the cont~min~nt by 25 sorption or by ion ex~h~n~e. A variety of these deto-x-icant particulates areknown in the art. A common one is carbon or graphite. Activated carbon is commonly employed for this purpose. A common concern with the use of such particulates is stated in Clark, U.S. 4,048,064, patented Sept. 13, 1977, at col.1, lines 8-23, as follows:

133~872 71746-4 "Whlle the technique is initially very effective, such prevlous attempts at hemoperfuslon have been plagued by very high losses of white cells and platelets (cite) as well as clotting, sludging, and channeling of blood in the column. The column then becomes ineffective and the patient suffers thromobocytopenia. Further, flne de-toxicant particles tend to be released into the blood stream to become emboll in blood vessels and organs such as the lungs, spleen, and kidneys (cite)."
Clark describes the use of a heparln loaded polymer coatlng of the carbon partlcles, preferably activated carbon, to provlde a semlpermeable coating on the partlcles. A mass of the coated partlcles ls then placed in a nylon mesh sack and the sack is placed in a container that has an inlet on one end and an out-let on the other. The blood is then fed to the container and passed through the bed of coated activated carbon particles where it penetrates the polymer coating, obtains heparln treatment and the contamlnatlon ls removed by adsorptlon.
Winchester, et al. Clinical Toxicoloqy, 17 (4), pp.
557 - 569 (1980) describe the use of a variety of sorbents for the detoxification by hemoperfusion of contaminated blood where the contamlnatlon was a chemotherapeutic agent (drug). The article shows that polymer coated and uncoated particulate detoxicants can be used to materially reduce the contamination of the blood. Like Clark, acrylic hydrogels were used to coat the particles. Non-lonlc exchange reslns are also descrlbed ln the artlcle. The authors found that a certaln pyrolized resin was an effective adsorbant.

~ 24 133~872 71746-4 Needless to say, modiflcatlons of the separation canl-sters suggested by Clark and Winchester, et al. wlll be dictated by the blood flow rate and the degree of drug contamlnation. If the blood flow rate ls higher ln the separatlon canlster contain-ing the bed or mass of the sorbents than the particle strength of the sorbent, then the techniques of treating the sorbents or operating the separatlon should be altered. For example, lf the sorbent breaks up ln the course of hemoperfuslon, then the sorbent should be larger and the flow rates of the contamlnated blood lnto and through the bed should be lower. Another approach to such a problem is to eliminate any leachables such as heparin from the polymer coating. This will make the coating more resistant to breakup.

~ 24a 1333~72 The pr jlem may also be helped by the use of stronger coatings which means the u~e of, e.g., a slightly more crosslinked pol~mer than those conventionally employed in the art for this purpose.

Hemodialysis has been previously employed .n cancer chemotherapy, 5 see Galletti, Portocaval Hemofiltration During The Anhepatic Phase In Isolated Liver Perfusion, Trans. Amer. Soc. Artif. Int. Organs, vol. XII, pp.
20 - 24, 1966, and Winchester, et al., Dialysis and hemoperfusion of poisons and drugs, Trans. Amer. Soc. Artif. Int. Organs, vol. XXIII, pp. 762 - 842.

Hemofiltration is a well defined technology and is characterized in a 10 number of texts. It involves the filtration from the cont~rnin~ted blood of the antineoplastic agent through membrane walls. -Details of the process and the apparatus used in effecting the process are described in inter alias Malchesky, Membrane Plasma Separation: Critical Issues, Therapeutic Apheresis: A
Critical Look, edited by Y. Nose', P. S. Malchesky, ~- ld J. W. Smith, ISAO Press, 15 No. 304, pp. 93 - 101, Cleveland, Ohio U.S.A., 1984; Vassilieff, et al., Plas-mapheresis Between a Rotating Truncated Cone and a Microporous Plate, Therapeutic Apheresis: A Critical Look, edited by Y. Nos~, P. S. Malchesky, and J. W. Smith, ISAO Press, No. 304, pp. 102 - 114, Cleveland, Ohio U.S.A., 1984;
R~ff, et al., Influence of Geometric Parameters on Filtration Flux in Plasma 20 Filters, Therapeutic Apheresis: A Critical L,ookj-edited by Y. Nose, P. S.
Malchesky, and J. W. Smith, ISAO Press, No. 304, pp. 115 - 121, Cleveland, Ohio U.S.A., 1984; Koga, et al., Investigation of the Clinical Properties of Various Filters for Double and Triple Filtration Plasmapheresis, Therapeutic A~heresis: A Critical Look, edited by Y. Nose', P. S. Malchesky, and J. W. Smith, 25 ISAO Press, No. 304, pp. 171 - 175, Cleveland, Ohio U.S.A., 1984; Tani, et al., New Anticancer Treatment by Hemoperfusion with Endotoxin Immobilized Fiber, Therapeutic Apheresis: A Critical Look, edited by Y. Nos~, P. S.
Malchesly, and J. W. Smith, ISAO Press, No. 304, pp. 202 - 207, Cleveland, Ohio U.S~, 1984; Fabbri, et al., Twelve-Hour Hemoperfusion on Activated 30 Coated Charcoal with Heparin and Prostacyclin in Healthy Rabbits.
Therapeutic Apher~sis: A Critical Look, edited by Y. Nosè, P. S. Malchesky, and J. W. Smith, ISAO Press, No. 304, pp. 208 - 216, Cleveland, Ohio U.S.A., 1984;

~ 8~75 - 74 and Gelfand, et al., Extracorporeal Induction of In Vivo Suupressor Cell rredominance bY Plasmaleukapherisis: An Alternative to CYclosporin in Renal TransDlantstion, Therapeutic Apheresis: A Crit~cal Look, edited by Y. Nose', P.
S. Malchesky, and J. W. Smi~h, ISAO Press, No. 304, pp. 217 - 240, Cleveland, Ohio U.S.A., 1984. A de~aled review of the subject Or hemorlltration cnn l)e found in Henne, et al., Membrane Technolo~ for Plasmapheresis, ~lasrna Separation and Plasma Fractionation, pp. 164 - 179 (Karger, Basel 1983).

An excellent overview Or hemofilltrntion nnd hemodialysis is presen~e(l in ~orton, et al., Continuous arteriovenous hemofiltration: An alternative to hemodial~sis, Arnerican Journal of Hospital P11arma~y, vol. 45, June 1988, pp.
1361 - 1368. The use of hemodialysis and hemoperfusion to remove antineop-lastic agents i~ described in Kamidono, et al., A Fundemental StudY Of Re~onal ChemotherapY Given BY Intraarterial Infusion With C~ncomitant Hemodialysis And Hemoperfusion. Inuestigatiue Urology, vol. 19, No. 3, pp. 176 - 178, 1981.

Hemofiltration and hemodialysis can be carried out with a renal~
Hemofiltration System, sold by renal systems, a division of Minntech 20 Corporation, Minneapolis, Mn 55441.

The chemical, physical or immunologic means for precipitation of the chemotherapeutic agent or the immunomodulating biologicals include separa-tion through hemoadsorbtion using antibodies or biological ligands or molecules able to render them nontoxic, and/or to clear the blood of the antineoplastic agent and allow the re-administration of the patient's own detoxified blood. These systems have enjoyed extensive consideration in the art and descriptions of them may be found in the foUowing publications:
Pineda, Method For Selective Removal Of Plasma Constituents. Therapeutic ,~lpheresis and Plasma Perfusion, pp. 361 - 373, 1982, Alan R Lis~, Inc., 150 30 Fifth Avenue, New York, NY 10011; Saal, et al., Extracorporeal Modification 0f Plasma And Whole Blood, Therapeutic Apheresis and Plasma Perfusion, pp.
375 - 384, 1982, Alan ~ Liss, Inc., 150 Fifth Avenue, New York, NY 10011; and Messerschmidt, et al., The Use or Protein-A In The Trea~ent or Malignanc~:
Rationa~e And The NCI ExPerience, Therapeulic Apheres~s a~d ~'lasma Perfusion, pp. 385 - 390, 1982, Alan ~ Liss, Inc., 150 Fifth Avenue, Neu York, NY 10011.

The pressure Or the blood af~er detoxi~lcation in the extracorporeal circuit is measured in the tubing. The pressure in the tubing must be main-10 tained above syslemic venous pressure. The dcloxirled blood is relurrle(l lothe body through the subclavian veins v~a a catheter percutaneously placed.
Alternatively, the blood may be returned throug~i a return lumen that traverses the double balloon catheter and delivers the detoxified blood to ~he right atrium or the superior vena cava.

Chemotherapy of the Organ The prior art illustrates a variety of procedures for supplying a chemotherapuetic agent to an organ containing a malignant tumor. These procedures may be used in the practice of the process of this invention. For example, using the Seldinger technique, a perfusion catheter can be intro-20 duced into the femoral artery. With nuoroscopy and standard arteriographictechniques, the catheter can be manipulated into the hepatic artery or, if the tumor is localized, into the branch of the hepatic artery that supplies the tumor. Arterial blood reaches normal liver cells from two sources, the hepatic artery and the portal vein. In the normal individual, 75% of the volume of the arterial blood that perfuses normal liver cells is delivered by the portal vein,while the hepatic artery supplies only about 25%. The catheter can be sutured to the skin of the groin to maintain its position. This catheter can be used rorinfusion of suitable anti-cancer agents directly into the artery that supplies the tumor. This procedure can be repeated as often as it would be clinically useful.
30Illustrntive of suitable chemotherapuetic agents for use in the practice of this invention are Adriamycin (doxorubicin), fluorinated pyrmidines (5-FU

i -~

1333g72 BGHOOl or noxuridine (FUDR)), cisplatin, Mitomycin C, cyclophosparnide, methot-rexate, vincristine, Bleomycin, FAMT, and any other anti-cancer agent, As pointed out above, the invention may be employed to effect treatment of organs with biologicals (immunomodulators) as part of a cancer therapy.
5 Illustrative immunomodulating biologicals suitable for use in the invention are alpha interferon, beta interferon, ~nma interferon, interleukin-2, interleukin-3, tumor necrosis factor, granulocyte-macrophage colony-stimulating factors, and the like.

Selective arteriography can be performed through the arterial 10 infusion c~theter prior to drug infusion and again prior to removal of the catheter to demonstrate the anatomic distribution of the supplying arteries and to evaluate any early changes in the vascular pattern of the tumor or the liver. In patients with accessoIy blood supply to the liver from either the superior mesenteric artery or the left gastric artery or both, the vessel which 15 supplies the main bulk of the tumor is the vessel that is infused. If more than one arte~y supplies large volumes of blood to the tumor, two infusion catheters may be used, one placed through each femoral artery, and dividing the infused dose of drug between the two catheters.

The arterial catheter can be removed immediately following post-20 infusion arteriography, and the vena cava catheter can be removed after thelast hepatic venous blood samples have been collected. Heparin can be given intravenously at least 15 minutes before the extracorporeal circuit is opened and discontinued or reversed with protamine shortly after deflation of the balloons but before removal of the venous catheter.

Liver function tests will be measured before therapy, daily for three days following therapy, and then at least monthly for at least 3 months to evaluate the possibility of functional alterations of the liver, either as a result of the perfusion technique, drug toxicity or growth of the tumor.

The process of the invention can be used repeatedly over periods of 30 months or years and as frequently ss desired, since the catheter should not BGHOOl produce permanent alteration of anatomy at the puncture site, at the site of arterial infusion, or at the site where the balloons are innated.

With respect to Figure 1, there is shown the featured components of the apparatus assembly of the invention used to practice the process of the 5 invention in relation to a human body 2. Liver 3 is supplied with cancer therapy drugs from syringe 4 through tubing leading to catheter 6 located in hepatic artery 5. The hepatic venous blood cont~inin~ anti-cancer concentra-tions of chemotherapuetic agent is passed via the hepatic veins 7 to the double balloon catheter located in rVc 1. The balloons of the double balloon catheter 10 are positioned central and peripheral of the hepatic veins 7. The cont~min~qted blood is passed through the double balloon catheter to tubing 17 to a point exterior to the body 2, thence to a pump 21 such as a Bio Medicus BP-50 Bio-Pump having a priming volume of 48 ml, cont~inin~ two rotator cones and providing a m~imum flow rate of 5 liters per minute. Pump 21 moves the 15 blood through the extracorporeal circuit at relatively constant low pressure,the object being to avoid raising or lowering the fluid pressure of the total circuit r~n~in~ from the hepatic veins through the return to the body. The cont~min~ted blood is transported through tubing 41 into detoxification zone 43, which in this case is a hemoperfusion cartridge cont~inin~ activated carbon.20 Suitable cartridge systems are obtainable from Clark Research and Develop-ment, Inc., New Orleans, I~ 70121 and from Garnbro Dialysatoren KG, d-7450 Hechingen, Federal Republic of Germany AUT 224 (sold under the trademark of ADSORBA~). The detoxified blood is passed through tube 44 to effect infusion through the subclavian vein (not shown) by standard procedures in 25 the art.

With respect to Figure 2, there is shown the relationship of inferior vena cava 1 to liver 3, hepatic veins 7 and portal veins 5. The hepatic artery is not shown in the drawing Double balloon catheter 9 comprises central balloon 11 and peripheral balloon 12, each in juxtaposition to cylindrical fenestration 30 zone 8. Zone 8 contains fenestrations 13 sufficient in total area to allow the complete removal of the hepatic venous flow into the catheter 9. The hollow interior (main lumen) of catheter 9 is of sufficient size to completely remove the blood from the hepatic veins without elevating hepatic back pressure.

133387~

BCHOOl Catheter 9 is provided with channel 15 that is used to inject flu.l into the balloons 11 and 12 for inflation or to ithdraw fluids for deflation. The ~enous now is passed through catheter 9 nto openly connected lube 17. Tube 17 may be interrupted by a pressure monitor the sarne as assembly A, discussed below, 5 that is later provided in the extracorporeal circuit. Tube 17 msy connect directly with pump 21 or to Y-fitting 19, as shown. Also connected to Y-fitting 19 is ancillary feed s~stem B comprising tube 23, ~J-fitting 25, and multiple IVspikes 29 and 33 each connected to tubes 30 and 31 respectively, and each is provided with a clarnp, 27 and 28, respectively. These lines can be used for the10 introduction of medications as required.

Pump 21 is a smooth rotator pump design and a particularly desirable pump is a Bio Medicus BP-50 Bio-Pump having a priming volume of 48 ml, cont~inir g two rotator cones and providing a m~ximum flow rate of 5 liters per minute. The cont~min~ted blood is gently pu~hed between the smooth 15 rotators 37 in zones 35 and issued from the pump through port 39 into tube 41.
Tube 41 is connected to cartridge or canister 43 cont~inine a meshed sack of activated carbon particles coated with an acrylic resin cont~ining heparin, see Clark, supra. The outflow from cartridge 43 is fed to tube 45 and then to tube 47 that is connected to pressure monitoring assembly ~ Pressure monitoring 20 assembly A comprises a pressure monitor gauge 55 connected to fluid membrane vessel 53 that contains a thin membrasre that separates the gauge 55 from the blood in vessel 53 and responds to the fluid pressure of the blood in vessel 53. That response is read by the gauge. Vessel 53 is connected to tubing 57, that is connected to stopcock 52. Stopcock 52 is connected to 25 flexible tubing 59 that in turn is connected to stopcock 51, the latter secured in fitting 49.

Blood from tubing 47 is passed to Y-connector 63 via tubing 61, then to tubings 65 and 67. Tubings 65 and 67 are each connected to catheter 69 and another catheter (connected to tube 65) not shown. These catheters are 30 provided for returning the purified blood to the subclavian veins.

In Figure 3, there is shown a double balloon catheter design that can have up to a 24 French (Fr) O.D. Zone 100 is provided with slotted fenestra-BGHOOl tions 104 in the solid plastic tubing 102. The open end 118 termin~tes the catheter. End 118 is tapered to the caliber of an angiographic guide wire that will, under fluoroscope control, allow the catheter to be advanced from the femorsl vein to the proper location in the inferior vena cava without risk of 5 injury to the interior of the vessels. Appropriate guide wires may be, for example, 0.035, 0.038, or 0.045 inch in diameter. During treatment, the catheter end hole is closed using a standard angiographic apparatus (tip-occluding wire), that consists of a thin wire long enough to traver~e the lengthof the catheter at the end of which is a stainless steel bead just large enough to 10 obstruct the catheter'q end-hole when advanced into it (similPr to a metal stopper that closes the outlet from a sink when advanced).

Alternatively, the end hole can be made 7-lZ Fr in diameter in order to accommodate a return catheter. The return catheter can be used to return treated blood to the systemic circulation. The return catheter is advanced 15 over a guide wire through the main lumen of the double balloon catheter and through the end hole 118 into the right atrium or superior vena cava. The return catheter can be made to graduslly taper its O.D. by decreasing its wall thickness, leaving the I.D. constant, since the location of the tip of the return catheter is not critical. The length over which the catheter tapers is arbitrary.
20 The taper is constructed so that the tip of the catheter is its narrowest O.D.
and the O.D. increases toward the femoral vein. As this return catheter is advanced through the lumen of the main catheter the tip easily passes through the end hole 118 of the double balloon catheter. The tapered end of the return catheter is advanced until it obstructs the end hole 118, preventing 25 systemic blood from entering the double balloon catheter when the balloons are inflated but lesving sn open lumen through the return catheter to return blood beyond the isolated venous seg~nent without mi~ing with cont~min~ted blood.

The catheter tubing (body) can be made of a variety of plastic 30 materials such as poly~roF~lene, polyethylene, polyvinylchloride, ethylene vinylacetate copolymers, polytetrafluoroethylene, polyurethane, and the like.
A favorable plastic comhin~tion for catheters cont~inin~ a return lu~nen are a homogeneous mixture of high density polyethylene and linear low density BGHOOI
polyethylene. That combination gives favorable stiffness at arnbient condi-tions and allows the use of especially thin wall thicknesses. When th~ surface of the catheter is made of a p.astic that is dif~lcult to bond with a balloon, the plastic may be treated first by one or more of a number of well known methods 5 that make bonding possible. The methods include plasma treatment, ozone treatment, and the like. Balloons 110 and 114 may be made from a plurality of elastomeric materials such as latex rubber, polyurethanes, spandex type polyurethanes, EPDM rubber, and the like. The balloons are typically ad-hesively bonded at sheath surfaces 108 and 112, respectively. A wide ariety of 10 a&esives may be employed. Polyacrylonitrile type adhesives, rubber latex adhesives and the like may be used to secure the balloon to the sheath sur-faces 108 and 112.

With respect to Figure 4, there is shown a cross section of a typical catheter design such as that shown in Figure q. The interior of the catheter 15 contains main lumen 120 and 4 additional lumina 124 molded into the outer wall 122. The additional lumina can be used for the various functions described above.

With respect to Figure 5, there is shown a cross section of another catheter design such as that shown in Figure 3 but cont~inin~ only three 20 ll-min~ The interior of the catheter contains main lumen 130 and two supplementary lumina 131 molded into segrnent 133 of wall 135. The sup-plementary lumina can be used for the various functions described above.

With respect to Figure 6, there is shown a double balloon catheter design which can have an outside diameter of 24 French such as in the 25 fenestration zone 140 and an inside diameter of less than 22 Fr. Zone 140 is provided with slotted fenestrations 141 in the plastic tubing 142. With respect to Figure 7, it is a cross sectional view of another typical catheter design showing a main lumen 150 and 3 supplemental lumina 151. Figure 8 provides a more detailed schematic cross sectional side view of a typical double balloon 30 catheter 161. .In this depiction, the catheter sidewall 163 iS penetrated by a plurality of fenestrations 165. The main lumen 169 contains at its periphery supplemental lumina 170,171 and 173. Supplemental lumen 170 can be used 13338~2 saHool to accommodate a guidewire, supplemental lumen 171 can be used to accom-modate a pressure monitor, and supplemental lumen 173 is used to supply fluid to the balloons 166 and 167 through openings 175 and 177.

Though this invention has been described with emphasis on the 5 treatment of cancer, it is quite apparent that the invention has broader application. The invention is useful for the treatment of any organ in which the treating agent would cause toxological ef~ects if it entered the body's general circulation. For example, the invention could be applied to the treatment of infectious deseases of organs such as fungal desea~es. A specific 10 illustration would be the treatment of hepatic fungal infections with Am-photericen B. The procedures described above would be directly applicable to extracorporeal recovery of this agent and its isolation from entering the general circulation of the body during treatment of the liver with significant concentrations of this drug.

Therefore, the breadth of the invention encompasses the perfusing of a high concentration of an agent to treat an organ, such as anti-cancer agents through a body organ cont~ining a tumor, without their entering the body's general circulation, removing them from the organ with effluent blood and transporting the cont~min~ted blood to an extracorporeal circuit where the 20 blood is treated to remove the cont~rnin~tion, and returning the treated blood to the body. The process prevents toxic levels of the agents from entering the body's general circulation while delivering lethal doses of the agents to the tumor.

Claims (41)

1. An apparatus for perfusing a high concentration of an agent through a body organ requiring treatment without contaminating a body's general circulation, said apparatus comprising:
- means for removing the agent from the organ with effluent blood;
- means for transporting the contaminated blood to an extracorporeal circuit;
- means for treating the blood in the extracorporeal circuit to remove the contamination; and - means to return the treated blood to the body.
2. The apparatus according to claim 1 wherein said agent is an anti-cancer agent.
3. The apparatus of claim 2 which comprises - means to perfuse an anti-cancer agent to a tumor, - means to collect and contain the contaminated blood emanating from the tumor without general circulation of the contaminated blood to the body, - means to transport the contaminated blood from the body to an extracorporeal treatment system, - means to remove the anti-cancer agent from the blood in the extracorporeal treatment system, and - means to return the treated blood to the body.
4. An apparatus to treat an organ site tumor which comprises a. means to expose a tumor in a body organ to one or more anti-cancer agents in higher than usual concentrations, b. means to remove from the organ effluent blood contaminated with the agent provided to the organ, without systemic exposure to the body, c. means to pass the effluent blood from tributary veins in the organ into a larger vein in which has been provided a catheter containing i. at least one inflatable balloon provided to obstruct passage of the effluent blood to the heart and ii. an avenue in the catheter sufficient to accommodate the volume of effluent blood traversing the tributary veins;
d. means to transport the contaminated effluent blood through the catheter and thence from the body into an extracorporeal circuit, e. means to detoxify the blood in the extracorporeal circuit, and f. means to return the detoxified blood to the body.
5. An apparatus for treating organ site tumors which comprises a. means to expose a tumor in a body organ to one or more antineoplastic drug and biological concentrations, b. means to remove from the organ effluent blood contaminated with the drug and biological provided to the organ, without systemic exposure to the body, c. means to pass the effluent blood to tributary veins emanating from the organ into a larger vein in which has been provided a catheter containing i. spaced-apart inflatable balloons provided to obstruct the large vein above and below said tributary veins and ii. an avenue in the catheter between the balloons sufficient to accommodate the volume of effluent blood being transported through the tributary vein, d. means to transport the contaminated effluent blood through the catheter and thence from the body into an extracorporeal circuit, e. means to detoxify the blood in the extracorporeal circuit and f. means to return the detoxified blood to the body.
6. The apparatus of claim 4 or 5 wherein the avenue comprises a plurality of holes in the catheter loading to the main lumen thereof.
7. The apparatus of claim 4 or 5 wherein the avenue comprises a single opening in the catheter.
8. The apparatus of claim 4 or 5 wherein the avenue contains a return lumen for returning detoxified blood to the body.
9. The apparatus of claim 6 wherein the avenue contains a return lumen for returning detoxified blood to the body.
10. The apparatus of claim 7 wherein the avenue contains a return lumen for returning detoxified blood to the body.
11. The apparatus of claim 2 wherein said means to return the blood to the body is an appropriately large caliber vein.
12. The apparatus of claim 11 wherein the large caliber vein is one or more of the subclavian veins.
13. Apparatus to remove an antineoplastic agent from hepatic venous blood before entering the systemic circulation thereby permitting safe infusion of anti-cancer levels of antineoplastic agent into the hepatic artery for treatment of the liver which apparatus comprises - means to perfuse the antineoplastic agent to the liver, - means to collect and contain the contaminated blood from the liver without general circulation of the contaminated blood to the remainder of the body, - means to transport the contaminated blood from the body to an extracorporeal treatment system for the contaminated blood, - an extracorporeal treatment system for removing the anti-neoplastic agent from the blood, and - means to return the treated blood to the body.
14. The apparatus of claim 1 wherein the organ is treated for an infectious disease.
15. The apparatus of claim 14 wherein the disease is a hepatic fungal infection.
16. The apparatus of claim 15 wherein the treatment involves the use of Amphotericin B.
17. A catheter adapted for percutaneous insertion into a vein or artery, comprising: a plastic tube defining a main lumen for outflowing blood, two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to the cranial end of said plastic tube, and said balloons, when inflated, having a size sufficient to block the flow of blood in a vein or artery into which said catheter is designed to be inserted; fenestrations in said plastic tube between said balloons to said main lumen; and second and third lumina with said plastic tube, said second lumen connecting to the caudal of said balloons and said third lumen connecting to the cranial of said balloons for effecting inflation or deflation of said balloons, the cranial end of said plastic tube being closed to any appreciable inflow of blood.
18. The catheter of claim 17 wherein said cranial end is tapered to a caliber of an angiographic guide wire.
19. The catheter of claim 17 wherein there is a fourth lumen within said plastic tube adapted to accommodate an angiographic guide wire.
20. The catheter of claim 17 wherein said second and third lumina connect and are common to the interiors of said balloons.
21. The catheter of claim 17 wherein said second and third lumina lie within the wall of said plastic tube.
22. The catheter of claim 17 wherein said plastic tube is adapted to carry a smaller return catheter and said cranial end tapers to fit about said return catheter.
23. The catheter of claim 17 wherein said plastic tube contains an additional lumen adapted to monitor blood pressure.
24. The catheter of claim 17 wherein the length of said plastic tube containing said fenestrations is in the range of nine to ten centimeters and said plastic tube has a maximum outside diameter of 24 French.
25. The catheter of claim 17 wherein the caudal end is long enough to extend through the skin of the patient to the outside of the body and terminates in a Luer fitting and a cut-off valve.
26. The catheter of claim 17 wherein said balloons are spaced apart a distance sufficient to bridge the hepatic veins when said catheter is inserted in the inferior vena cava and said fenestrations are sufficient to remove blood issuing from said hepatic veins without elevating blood pressure.
27. A kit for removing a treating agent from blood perfused through a body organ of a patient, comprising:
a catheter for isolating and removing the blood issuing from said body organ and containing said treating agent;
a detoxification means for treating said blood so removed to remove said treating agent; and return catheter means for returning the blood so treated to said patient;
said catheter comprising:
a plastic tube defining a main lumen for outflowing blood, two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to the cranial end of said plastic tube and said balloons, when inflated, having a size sufficient to block the flow of blood in a vein into which said catheter is designed to be inserted;
fenestrations in said plastic tube between said balloons to said lumen and second and third lumina within said plastic tube, said second lumen connecting to the caudal of said balloons and said third lumen connecting to the cranial of said balloons for effecting inflation or deflation of said balloons, the cranial end of said plastic tube being closed to any appreciable inflow of blood.
28. The kit of claim 27 wherein said kit is effective for a treatment selected from the group consisting of hemoperfusion, hemodialysis, hemofiltration and hemoabsorption.
29. The kit of claim 27 wherein said return catheter fits within said plastic tube and said cranial end tapers to fit thereabout such that the treated blood is returned past the cranial balloon and said kit includes arterial injection means for introducing said treating agent into an artery leading to said body organ.
30. The kit of claim 27 wherein said body organ is the liver and said treating agent is an anti-cancer agent.
31. The kit of claim 27, including, in addition, an angiographic guide wire and said cranial end is tapered to the caliber of said angiographic guide wire.
32. Apparatus for isolation and treatment of a portion of the body comprising a first catheter adapted for percutaneous insertion into a vein or artery, comprising (a) a plastic tube having a cranial end and a caudal end, said plastic tube defining a main lumen for outflowing blood; two balloons, fixedly spaced apart about said plastic tube and bonded thereto for inflation thereabout, one being contiguous to said cranial end, and said balloons, when inflated, having a size sufficient to block the flow of blood in a vein or artery into which said catheter is designed to be inserted; fenestrations in said plastic tube between said balloons to said main lumen, second and third lumina within said plastic tube, said second lumen connecting to one of said balloons and said third lumen connecting to the other of said balloons for effecting inflation or deflation of said balloons, said cranial end of said plastic tube being closed to any appreciable inflow of blood and (b) a second catheter for returning blood removed through said main lumen to the patient.
33. The apparatus of claim 32 wherein said cranial end is tapered to a caliber of an angiographic guide wire.
34. The apparatus of claim 33 wherein said plastic tube has a fourth lumen adapted to accommodate an angiographic guide wire.
35. The apparatus of claim 32 wherein said second and third lumina connect and are common to the interiors of said balloons.
36. The apparatus of claim 32 wherein said second and third lumina lie within the wall of said plastic tube.
37. The apparatus of claim 32 wherein said plastic tube contains an additional lumen adapted to monitor blood pressure.
38. The apparatus of claim 32 wherein the length of said plastic tube obtaining said fenestrations is in the range of nine to ten centimeters and said plastic tube has a maximum outside diameter of 24 French.
39. The apparatus of claim 32 wherein said caudal end is long enough to extend through the skin of the patient to the outside of the body and terminates in a Luer fitting and a shut-off valve.
40. The apparatus of claim 32 wherein said balloons are spaced apart a distance sufficient to bridge the hepatic veins when said catheter is inserted in the inferior vena cava and said fenestrations are sufficient to remove blood issuing from said hepatic veins without elevating blood pressure.
41. A use of an apparatus according to any one of claims 1 to 5, 9 to 16 and 32 to 40 to treat a body organ without contaminating a body's general circulation.
CA000612992A 1988-10-21 1989-09-25 Cancer treatment Expired - Lifetime CA1333872C (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US260,623 1981-05-05
US07/260,623 US5069662A (en) 1988-10-21 1988-10-21 Cancer treatment

Publications (1)

Publication Number Publication Date
CA1333872C true CA1333872C (en) 1995-01-10

Family

ID=22989916

Family Applications (1)

Application Number Title Priority Date Filing Date
CA000612992A Expired - Lifetime CA1333872C (en) 1988-10-21 1989-09-25 Cancer treatment

Country Status (7)

Country Link
US (2) US5069662A (en)
EP (1) EP0364799B1 (en)
JP (1) JP2831056B2 (en)
AT (1) ATE121952T1 (en)
CA (1) CA1333872C (en)
DE (1) DE68922456T2 (en)
ES (1) ES2075022T3 (en)

Families Citing this family (211)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69034042T2 (en) * 1989-08-02 2003-07-17 Mitra Medical Technology Ab Lu SYSTEM FOR USE IN A METHOD FOR THERAPEUTIC AND DIAGNOSTIC TREATMENT
US5423754A (en) * 1990-09-20 1995-06-13 Scimed Life Systems, Inc. Intravascular catheter
SE9100142L (en) 1991-01-17 1992-07-18 Bengt Sandberg A METHOD AND A SYSTEM FOR PREPARING VIVO REDUCTION OF DIAGNOSTIC AND / OR THERAPEUTIC SUBSTANCES BY EXTRACORAL REMOVAL AND THE USE OF THESE SUBSTANCES FOR THIS PURPOSE
US5454788A (en) * 1991-04-24 1995-10-03 Baxter International Inc. Exchangeable integrated-wire balloon catheter
DE69224636T2 (en) * 1991-04-24 1998-11-05 Advanced Cardiovascular System INTERCHANGEABLE BALLOON CATHETER WITH INTEGRATED GUIDE WIRE
US6482171B1 (en) 1991-07-16 2002-11-19 Heartport, Inc. Multi-lumen catheter
US5584803A (en) 1991-07-16 1996-12-17 Heartport, Inc. System for cardiac procedures
US5769812A (en) 1991-07-16 1998-06-23 Heartport, Inc. System for cardiac procedures
CA2074304C (en) * 1991-08-02 1996-11-26 Cyril J. Schweich, Jr. Drug delivery catheter
US5308342A (en) * 1991-08-07 1994-05-03 Target Therapeutics, Inc. Variable stiffness catheter
US6224619B1 (en) 1991-12-17 2001-05-01 Heartport, Inc. Blood vessel occlusion trocar having size and shape varying insertion body
US5257979A (en) * 1992-07-27 1993-11-02 Ravindar Jagpal Instrument for catheterization
US5318519A (en) * 1992-08-19 1994-06-07 Wilk Peter J Method and apparatus for supplying nutrition
US5730722A (en) * 1992-08-19 1998-03-24 Wilk; Peter J. Method and apparatus for supplying a medical treatment composition to a patient
US5544651A (en) * 1992-09-08 1996-08-13 Wilk; Peter J. Medical system and associated method for automatic treatment
AU7676894A (en) * 1993-08-27 1995-03-21 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Convection-enhanced drug delivery
US5695479A (en) * 1993-11-01 1997-12-09 Jagpal; Ravindar Instrument, system, kit and method for catheterization procedures
ES2077519B1 (en) * 1993-11-22 1996-07-01 Fernandez De Lomana Euge Anaya INTRAAORTIC CATHETER FOR RENAL PERFUSION AND CONSERVATION.
US7008395B1 (en) * 1994-04-04 2006-03-07 Wake Forset University Health Sciences Multi-lumen catheter system used in a blood treatment process
AU2379495A (en) * 1994-04-04 1995-10-23 Brian W. Loggie Multi-lumen catheter system
US5478309A (en) 1994-05-27 1995-12-26 William P. Sweezer, Jr. Catheter system and method for providing cardiopulmonary bypass pump support during heart surgery
US5681278A (en) * 1994-06-23 1997-10-28 Cormedics Corp. Coronary vasculature treatment method
DE19514638C2 (en) * 1995-04-20 1998-06-04 Peter Dr Med Boekstegers Device for the selective suction and retroinfusion of a fluid from or into body veins controlled by venous pressure
US5900433A (en) * 1995-06-23 1999-05-04 Cormedics Corp. Vascular treatment method and apparatus
US5736129A (en) * 1995-11-17 1998-04-07 Medenica; Rajko D. Flow cytometric pharmacosensitivity assay and method of cancer treatment
US5868703A (en) * 1996-04-10 1999-02-09 Endoscopic Technologies, Inc. Multichannel catheter
US6821265B1 (en) 1996-04-10 2004-11-23 Endoscopic Technologies, Inc. Multichannel catheter
JP3526690B2 (en) * 1996-04-26 2004-05-17 富雄 太田 Extracorporeal circulation device for separate cooling method
US5876366A (en) * 1996-07-22 1999-03-02 Dykstra; Todd M. Kidney dialysis method and device
US6186146B1 (en) * 1996-08-30 2001-02-13 Delcath Systems Inc Cancer treatment method
US5879320A (en) * 1996-12-23 1999-03-09 Cazenave; Craig Richard Implantable vascular device
US5879321A (en) * 1997-01-22 1999-03-09 The University Of Kentucky Research Foundation Portocaval-right atrial shunt
CA2278698C (en) * 1997-02-04 2007-11-20 Cook Urological Inc. Suprapubic drainage catheter
US5755687A (en) 1997-04-01 1998-05-26 Heartport, Inc. Methods and devices for occluding a patient's ascending aorta
US5980481A (en) * 1997-05-08 1999-11-09 Transvivo, Inc. Method and apparatus for continuous peritoneal cascade dialysis and hemofiltration (CPCD/H)
EP0998565A1 (en) * 1997-07-11 2000-05-10 Introgene B.V. Interleukin-3 gene therapy for cancer
US5919163A (en) * 1997-07-14 1999-07-06 Delcath Systems, Inc. Catheter with slidable balloon
US5817046A (en) * 1997-07-14 1998-10-06 Delcath Systems, Inc. Apparatus and method for isolated pelvic perfusion
US7037316B2 (en) 1997-07-24 2006-05-02 Mcguckin Jr James F Rotational thrombectomy device
US6416487B1 (en) 1997-07-30 2002-07-09 Renal Tech International Llc Method of removing beta-2 microglobulin from blood
US20010041862A1 (en) 1997-09-02 2001-11-15 Morton G. Glickman Novel apparatus and method of treating a tumor in the extremity of a patient
US5897533A (en) * 1997-09-02 1999-04-27 Delcath Systems, Inc. Catheter flow and lateral movement controller
EP1024753B1 (en) * 1997-09-26 2003-04-09 Duke University Perfusion-occlusion catheter
UA56262C2 (en) 1997-10-09 2003-05-15 Орквіс Медікел Корпорейшн Extracardiac pumping system for supplementing blood circulation
US6889082B2 (en) 1997-10-09 2005-05-03 Orqis Medical Corporation Implantable heart assist system and method of applying same
US6610004B2 (en) 1997-10-09 2003-08-26 Orqis Medical Corporation Implantable heart assist system and method of applying same
US6390969B1 (en) 1997-10-09 2002-05-21 Orqis Medical Corporation Implantable heart assist system and method of applying same
US6387037B1 (en) 1997-10-09 2002-05-14 Orqis Medical Corporation Implantable heart assist system and method of applying same
US6699231B1 (en) 1997-12-31 2004-03-02 Heartport, Inc. Methods and apparatus for perfusion of isolated tissue structure
US6159178A (en) * 1998-01-23 2000-12-12 Heartport, Inc. Methods and devices for occluding the ascending aorta and maintaining circulation of oxygenated blood in the patient when the patient's heart is arrested
US6722370B1 (en) * 1998-07-17 2004-04-20 Corgentech, Inc. Delivery of a composition to the liver by utilizing the portal vein
US6461327B1 (en) * 1998-08-07 2002-10-08 Embol-X, Inc. Atrial isolator and method of use
JP2004135688A (en) 1998-11-12 2004-05-13 Yoshino Seiki:Kk Balloon catheter for blood vessel
JP3655197B2 (en) * 1998-11-19 2005-06-02 富雄 太田 Extracorporeal circulation device for separation temperature control method
US6749598B1 (en) * 1999-01-11 2004-06-15 Flowmedica, Inc. Apparatus and methods for treating congestive heart disease
US7481803B2 (en) 2000-11-28 2009-01-27 Flowmedica, Inc. Intra-aortic renal drug delivery catheter
US7780628B1 (en) 1999-01-11 2010-08-24 Angiodynamics, Inc. Apparatus and methods for treating congestive heart disease
EP2189213A1 (en) 1999-01-22 2010-05-26 Dow Global Technologies Inc. Method for producing a surface modified divinylbenzene resin having a hemocompatible coating
EP1157709B1 (en) * 1999-02-10 2012-05-30 Tomio Ohta Bloodless treating device
DE19917901C2 (en) * 1999-04-20 2001-06-28 Univ Eberhard Karls Catheter set for preserving the heart and internal organs for transplants
CA2369576C (en) 1999-04-23 2005-07-05 William Frederick Weitzel Extracorporeal circuit and related methods
AUPQ090499A0 (en) 1999-06-10 1999-07-01 Peters, William S Heart assist device and system
US6815170B1 (en) 1999-06-30 2004-11-09 John Wayne Cancer Institute Methods for lymph node identification
SE516215C2 (en) 1999-07-10 2001-12-03 Argmed Kb Perfusion system and pump device and control system for use in the perfusion system
US7892201B1 (en) * 1999-08-27 2011-02-22 Gore Enterprise Holdings, Inc. Balloon catheter and method of mounting same
US8414543B2 (en) 1999-10-22 2013-04-09 Rex Medical, L.P. Rotational thrombectomy wire with blocking device
AU2614901A (en) 1999-10-22 2001-04-30 Boston Scientific Corporation Double balloon thrombectomy catheter
IT1310933B1 (en) * 1999-11-16 2002-02-27 Luigi Benatti MULTIFUNCTIONAL MACHINE FOR LOCOREGIONAL MONITORING AND DITERAPY CONTROL IN ONCOLOGY.
US6485500B1 (en) * 2000-03-21 2002-11-26 Advanced Cardiovascular Systems, Inc. Emboli protection system
US6569146B1 (en) * 2000-08-18 2003-05-27 Scimed Life Systems, Inc. Method and apparatus for treating saphenous vein graft lesions
US6692458B2 (en) * 2000-12-19 2004-02-17 Edwards Lifesciences Corporation Intra-pericardial drug delivery device with multiple balloons and method for angiogenesis
US6554819B2 (en) 2001-01-09 2003-04-29 Mount Sinai School Of Medicine Of New York University Method and device for preventing contrast associated nephropathy
AU2002235462A1 (en) * 2001-01-23 2002-08-06 The Regents Of The University Of California Method and apparatus to remove substances from vessels of the heart and other parts of the body to minimize or avoid renal or other harm or dysfunction
US7914478B2 (en) * 2001-03-30 2011-03-29 Tatsuo Kumazaki Intrapelvic perfusion therapy with cancer therapeutic agent and apparatus for intrapelvic perfusion with cancer therapeutic agent
AUPR669001A0 (en) 2001-07-30 2001-08-23 Sunshine Heart Company Pty Ltd A fluid pressure generating means
WO2003028642A2 (en) * 2001-10-01 2003-04-10 Rhode Island Hospital, A Lifespan Partner Methods of inhibiting metastases
US7383073B1 (en) * 2001-10-16 2008-06-03 Z-Kat Inc. Digital minimally invasive surgery system
US6913610B2 (en) * 2001-10-16 2005-07-05 Granit Medical Innovations, Inc. Endoscopic retractor instrument and associated method
US20040053367A1 (en) * 2001-12-04 2004-03-18 Griffin Jennifer A. Lipid-associated molecules
DE20203840U1 (en) * 2002-03-09 2002-11-14 Brodersen Jens Sclerotherapy catheter for the obliteration of varicose veins in the leg area
ATE386568T1 (en) 2002-03-09 2008-03-15 Jens P Brodersen SCLEROSIZATION CATHETER FOR SCLEROSIZATION OF BLOOD VESSELS, PARTICULARLY VEINS
US6926725B2 (en) 2002-04-04 2005-08-09 Rex Medical, L.P. Thrombectomy device with multi-layered rotational wire
US20030199449A1 (en) * 2002-04-19 2003-10-23 Tarcha Peter J. Combination of ablation and controlled drug delivery for the treatment of cancer
US7166120B2 (en) * 2002-07-12 2007-01-23 Ev3 Inc. Catheter with occluding cuff
US6849062B2 (en) * 2002-08-23 2005-02-01 Medtronic Vascular, Inc. Catheter having a low-friction guidewire lumen and method of manufacture
US20040044300A1 (en) * 2002-09-03 2004-03-04 Donnie Rudd Method of replenishing cells damaged by treatment for cancer
US20040077985A1 (en) * 2002-09-03 2004-04-22 Donnie Rudd Method of replenishing cells damaged by treatment for cancer
US7063679B2 (en) * 2002-09-20 2006-06-20 Flowmedica, Inc. Intra-aortic renal delivery catheter
WO2004034767A2 (en) 2002-09-20 2004-04-29 Flowmedica, Inc. Catheter system for renal therapy
US7993325B2 (en) 2002-09-20 2011-08-09 Angio Dynamics, Inc. Renal infusion systems and methods
EP1585572A4 (en) 2002-09-20 2010-02-24 Flowmedica Inc Method and apparatus for intra aortic substance delivery to a branch vessel
AU2003276903A1 (en) 2002-09-20 2004-05-04 Flowmedica, Inc. Method and apparatus for selective material delivery via an intra-renal catheter
AU2002952691A0 (en) 2002-11-15 2002-11-28 Sunshine Heart Company Pty Ltd Heart assist device utilising aortic deformation
US20030130610A1 (en) * 2002-12-09 2003-07-10 Mager Larry F. Aortic balloon catheter with improved positioning and balloon stability
US20040144387A1 (en) * 2003-01-28 2004-07-29 David Amar Double-balloon endobronchial catheter for one lung isolation anesthesia and surgery
ITBO20030239A1 (en) 2003-04-18 2004-10-19 Bruno Pasquale Franco Nardo DEVICE FOR THE ARTERIALIZATION OF THE DOOR VEIN AND RELATED KIT FOR ITS APPLICATION ON THE BODY OF A PATIENT.
US7022097B2 (en) * 2003-05-09 2006-04-04 Morton Glickman Method for treating glandular diseases and malignancies
JP2006526464A (en) 2003-06-05 2006-11-24 フローメディカ,インコーポレイテッド System and method for performing bilateral intervention or diagnosis in a branched body lumen
US20060167437A1 (en) * 2003-06-17 2006-07-27 Flowmedica, Inc. Method and apparatus for intra aortic substance delivery to a branch vessel
EP1659970A4 (en) * 2003-08-05 2008-05-21 Flowmedica Inc Sytem and method for prevention of radiocontrast induced nephropathy
US20070073264A1 (en) 2003-09-26 2007-03-29 The Trustees Of The University Of Pennsylvania Methods, compositions and apparatus for delivering heterologous molecules to cells
US7862499B2 (en) 2003-10-30 2011-01-04 Sunshine Heart Company Pty Ltd Blood vessel wrap
AU2004284842B2 (en) 2003-10-31 2010-03-04 Nuwellis, Inc. Synchronisation control system
US7887478B2 (en) 2003-10-31 2011-02-15 Sunshine Heart Company Pty Ltd Percutaneous gas-line
CN1878581B (en) 2003-11-11 2010-07-07 阳光心脏有限公司 Actuator for a heart assist device
US7695491B2 (en) * 2003-12-01 2010-04-13 Ev3 Inc. Rapid exchange catheters with tandem lumens
US20070203445A1 (en) * 2004-02-26 2007-08-30 V-Kardia Pty Ltd Isolating cardiac circulation
WO2005091910A2 (en) * 2004-03-04 2005-10-06 Flowmedica, Inc. Sheath for use in peripheral interventions
GB0406728D0 (en) * 2004-03-25 2004-04-28 Hydrodynamic Gene Delivery Ltd Gene therapy
EP1750506A4 (en) 2004-05-14 2010-03-17 Flowmedica Inc Bi-lateral local renal delivery for treating congestive heart failure and for bnp therapy
US7445592B2 (en) 2004-06-10 2008-11-04 Orqis Medical Corporation Cannulae having reduced flow resistance
US20060015065A1 (en) * 2004-07-15 2006-01-19 Tatsuo Kumazaki Method of drug perfusion in paraaortic lymph node tumors, sheath for inserting catheter, and oxygenated blood perfusion apparatus
US7758541B2 (en) * 2004-08-17 2010-07-20 Boston Scientific Scimed, Inc. Targeted drug delivery device and method
US20060069323A1 (en) * 2004-09-24 2006-03-30 Flowmedica, Inc. Systems and methods for bi-lateral guidewire cannulation of branched body lumens
US20090054823A1 (en) * 2004-09-30 2009-02-26 The Trustees Of The University Of Pennsylvania Perfusion circuit and use therein in targeted delivery of macromolecules
WO2006055813A2 (en) 2004-11-16 2006-05-26 Medrad, Inc. Modeling of pharmaceutical propagation
EP2990073B1 (en) * 2004-11-24 2018-05-02 Bayer Healthcare LLC Devices and systems for fluid delivery
WO2006073870A2 (en) * 2004-12-30 2006-07-13 Reynaldo Calderon M D Computerized system for monitored retrograde perfusion of tumor sites
EP1846080A4 (en) * 2005-01-19 2010-05-19 Sentinel Group Llc Medical agent delivery system and method
US20070167913A1 (en) * 2005-10-11 2007-07-19 Flowmedica, Inc. Vascular sheath with variable lumen construction
US20090187158A1 (en) * 2005-10-20 2009-07-23 Richmond Frank M Conversion device
US20070173798A1 (en) * 2006-01-23 2007-07-26 Adams Mark L Minimally invasive methods for thermal treatment
PT1996273E (en) 2006-03-20 2009-09-23 Medical Device Works Nv Sa A device for delivering medical treatment
US7771401B2 (en) 2006-06-08 2010-08-10 Angiodynamics, Inc. Selective renal cannulation and infusion systems and methods
EP2054103B1 (en) 2006-08-21 2019-05-29 Sunshine Heart Company Pty Ltd An improved wrap for a heart assist device
US8858584B2 (en) * 2006-11-07 2014-10-14 Cook Medical Technologies Llc Emergency transection device
WO2008085421A2 (en) 2006-12-29 2008-07-17 Medrad, Inc. Patient-based parameter generation systems for medical injection procedures
CN100473432C (en) * 2006-12-30 2009-04-01 中国人民解放军第三军医大学第一附属医院 Liver segment positioning catheter
EP2104468A2 (en) * 2007-01-08 2009-09-30 Gross, Yossi In-situ filter
WO2008108996A1 (en) * 2007-03-01 2008-09-12 Lifeline Scientific, Inc. Perfusion regulation
US20080221551A1 (en) * 2007-03-09 2008-09-11 Flowmedica, Inc. Acute kidney injury treatment systems and methods
US7824357B2 (en) * 2007-06-11 2010-11-02 Ibrahim Rashid Al-Rashdan Method and system for preventing contrast associated nephropathy
CN103976736B (en) 2007-07-17 2017-01-11 拜耳医药保健有限责任公司 Devices and systems for determination of parameters for a procedure, for estimation of cardiopulmonary function and for fluid delivery
US20090105799A1 (en) * 2007-10-23 2009-04-23 Flowmedica, Inc. Renal assessment systems and methods
EP2211936B1 (en) 2007-11-07 2014-07-16 LANE, Rodney James Systems and devices for circulatory access
EP2244662B1 (en) * 2008-01-28 2021-09-29 Implantica Patent Ltd. Blood clot removal device and system
US8892182B2 (en) * 2008-02-08 2014-11-18 Ethicon, Inc. Double balloon isolation catheters and methods therefor
US20090281634A1 (en) * 2008-05-08 2009-11-12 The University Of Mississippi Medical Center Research Development Foundation Gut prosthetic
WO2009149179A2 (en) * 2008-06-03 2009-12-10 Aethlon Medical Inc. Enhanced antiviral therapy methods and devices
US20100041984A1 (en) * 2008-08-12 2010-02-18 James Edward Shapland Impedance sensing device and catheter system
US9050400B2 (en) * 2008-08-12 2015-06-09 Osprey Medical, Inc. Remote sensing catheter system and methods
EP2340076B1 (en) * 2008-09-15 2019-11-06 Aeeg Ab Medical device and system for temporary occlusion of an opening in a lumen of a body
EP2334366A4 (en) * 2008-09-16 2011-10-05 Intersect Partners Llc Device and methods for sampling prostate fluid
US8162879B2 (en) * 2008-09-22 2012-04-24 Tyco Healthcare Group Lp Double balloon catheter and methods for homogeneous drug delivery using the same
US9421330B2 (en) 2008-11-03 2016-08-23 Bayer Healthcare Llc Mitigation of contrast-induced nephropathy
US8758287B2 (en) * 2008-11-12 2014-06-24 Marv Enterprises, LLC Utilization of stents for the treatment of blood borne carcinomas
ES2368404B1 (en) * 2008-12-01 2012-09-24 Fundacio Clinic Per A La Recerca Biomedica PERCUTANEOUS CATHETER AND METHOD FOR MEASURING PRESSURE IN BLOOD VESSELS.
US9216386B2 (en) 2009-03-17 2015-12-22 Marv Enterprises, LLC Sequential extracorporeal treatment of bodily fluids
US20100284510A1 (en) * 2009-05-08 2010-11-11 Jeffrey Neitlich Imaging gallbladder using computed tomography
WO2011056181A1 (en) * 2009-11-09 2011-05-12 Delcath Systems, Inc. Systems and methods for homeostatically treating organ disease using local delivery of therapeutic agents
US10512761B2 (en) 2009-12-02 2019-12-24 Renovorx, Inc. Methods for delivery of therapeutic materials to treat pancreatic cancer
US9295816B2 (en) * 2009-12-09 2016-03-29 Osprey Medical, Inc. Catheter with distal and proximal ports
EP2531251B1 (en) 2010-02-02 2019-12-04 Nirva Medical, LLC Localized therapy delivery and local organ protection
US9308310B2 (en) 2010-02-02 2016-04-12 Nirva Medical, Llc Localized therapy delivery and local organ protection
CN103037929B (en) 2010-03-06 2015-08-26 新融合血管系统有限公司 Reclaim catheter kit
DE102010012050B4 (en) * 2010-03-19 2016-09-29 Fresenius Medical Care Deutschland Gmbh hemodialysis
CN102939117B (en) 2010-04-02 2015-12-02 阳光心脏有限公司 Combination heart assist system, method and apparatus
CA2803169C (en) 2010-06-24 2020-09-22 Medrad, Inc. Modeling of pharmaceutical propagation and parameter generation for injection protocols
US10583246B2 (en) * 2011-03-09 2020-03-10 H. Lee Moffitt Cancer Center And Research Institute, Inc. High flow rate isolated infusion for regional treatment of cancer and medical conditions
JP2012200573A (en) * 2011-03-28 2012-10-22 Kobe Univ Two-balloon catheter
WO2013020044A1 (en) 2011-08-03 2013-02-07 The Charlotte-Mecklenburg Hospital Authority D/B/A Carolinas Medical Center Treatment of fibrosis using microrna-19b
EP2741804B1 (en) * 2011-08-11 2017-04-26 Osprey Medical Inc. Systems and methods for limb treatment
US9707331B2 (en) 2011-11-07 2017-07-18 Delcath Systems, Inc. Apparatus for removing chemotherapy compounds from blood
US8251942B1 (en) * 2011-12-19 2012-08-28 Ibrahim Rashid Al-Rashdan System and method for removing blood containing contrast dye
WO2013128016A1 (en) * 2012-03-01 2013-09-06 Medical Device Works Nv Kit and devices for organ perfusion
US20150027950A1 (en) * 2012-03-27 2015-01-29 Marv Enterprises, LLC Treatment for atherosclerosis
JP2015128457A (en) * 2012-04-27 2015-07-16 テルモ株式会社 embolus discharge catheter
HUE056182T2 (en) 2012-05-14 2022-01-28 Bayer Healthcare Llc Systems and methods for determination of pharmaceutical fluid injection protocols based on x-ray tube voltage
US9108029B2 (en) 2012-05-21 2015-08-18 Nfusion Vascular Systems, Llc Recovery catheter assembly and method
US9332998B2 (en) 2012-08-13 2016-05-10 Covidien Lp Apparatus and methods for clot disruption and evacuation
US9332999B2 (en) 2012-08-13 2016-05-10 Covidien Lp Apparatus and methods for clot disruption and evacuation
US9541544B1 (en) * 2012-08-28 2017-01-10 Delcath Systems, Inc. Method of selecting chemotherapeutic agents for an isolated organ or regional therapy
US9872851B2 (en) 2012-12-12 2018-01-23 The Charlotte-Mecklenburg Hospital Authority Methods of treating portal hypertension
US9750718B2 (en) 2012-12-12 2017-09-05 The Charlotte-Mecklenburg Hospital Authority Methods of treating hepatic fibrosis and associated diseases by regulating Rev-ERB activity
CN104812336B (en) * 2012-12-21 2018-06-15 加利福尼亚大学董事会 Filter device and its correlation technique can be positioned in vivo
US9555379B2 (en) 2013-03-13 2017-01-31 Bayer Healthcare Llc Fluid path set with turbulent mixing chamber, backflow compensator
US8795219B1 (en) 2013-03-15 2014-08-05 Ibrahim Rashid Al-Rashdan System and method for removing blood containing contrast dye
US9901722B2 (en) 2014-06-01 2018-02-27 White Swell Medical Ltd System and method for treatment of pulmonary edema
US20170232238A1 (en) * 2014-08-06 2017-08-17 Edwars Litesciences Corporation Multi-lumen cannulae
EP3294369B1 (en) 2015-05-11 2021-09-01 White Swell Medical Ltd Systems for reducing pressure at an outflow of a duct
WO2017152036A1 (en) 2016-03-03 2017-09-08 Bayer Healthcare Llc System and method for improved fluid delivery in multi-fluid injector systems
US10857328B2 (en) 2018-01-16 2020-12-08 Daniel Ezra Walzman Bypass catheter
US10799092B2 (en) * 2016-09-19 2020-10-13 Covidien Lp System and method for cleansing segments of a luminal network
CA3042153A1 (en) 2016-11-01 2018-05-11 White Swell Medical Ltd Systems and methods for treatment of fluid overload
US10857335B2 (en) * 2017-02-13 2020-12-08 Daniel Ezra Walzman Temporary bypass balloon catheter
WO2018158636A1 (en) 2017-03-02 2018-09-07 White Swell Medical Ltd Systems and methods for reducing pressure at outflow of a duct
WO2018172848A2 (en) 2017-03-19 2018-09-27 White Swell Medical Ltd Methods and devices for reducing pressure
US10695543B2 (en) 2017-05-18 2020-06-30 Renovorx, Inc. Methods for treating cancerous tumors
US11052224B2 (en) 2017-05-18 2021-07-06 Renovorx, Inc. Methods for treating cancerous tumors
EP3634528B1 (en) 2017-06-07 2023-06-07 Shifamed Holdings, LLC Intravascular fluid movement devices, systems, and methods of use
AU2018326485B2 (en) 2017-08-31 2024-01-04 Bayer Healthcare Llc Injector pressure calibration system and method
EP3675930B1 (en) 2017-08-31 2024-01-17 Bayer Healthcare LLC Method for drive member position and fluid injector system mechanical calibration
CN110809482B (en) 2017-08-31 2023-03-07 拜耳医药保健有限公司 Fluid injector system volume compensation system and method
US11141535B2 (en) 2017-08-31 2021-10-12 Bayer Healthcare Llc Fluid path impedance assessment for improving fluid delivery performance
US11779702B2 (en) 2017-08-31 2023-10-10 Bayer Healthcare Llc Method for dynamic pressure control in a fluid injector system
CN111556763B (en) 2017-11-13 2023-09-01 施菲姆德控股有限责任公司 Intravascular fluid movement device and system
US10799674B2 (en) 2018-01-16 2020-10-13 Daniel Ezra Walzman Bypass catheter
US11596769B2 (en) 2018-01-16 2023-03-07 Daniel Ezra Walzman Bypass catheter
US10926061B2 (en) 2018-01-16 2021-02-23 Daniel Ezra Walzman Bypass catheter
US11006996B2 (en) 2018-01-16 2021-05-18 Daniel Ezra Walzman Torus balloon with energy emitters for intravascular lithotripsy
US11596438B2 (en) 2018-01-16 2023-03-07 Daniel Ezra Walzman Bypass catheter
CN108030996B (en) * 2018-01-19 2020-06-30 北京大学第三医院 Integrated biliary tract self-taking-off internal drainage device
JP7410034B2 (en) 2018-02-01 2024-01-09 シファメド・ホールディングス・エルエルシー Intravascular blood pump and methods of use and manufacture
US11660426B2 (en) 2019-02-26 2023-05-30 White Swell Medical Ltd Devices and methods for treating edema
US11931560B2 (en) 2019-02-26 2024-03-19 White Swell Medical Ltd Devices and methods for treating edema
US11793996B2 (en) 2019-02-26 2023-10-24 White Swell Medical Ltd Devices and methods for treating edema
US11724095B2 (en) 2019-02-26 2023-08-15 White Swell Medical Ltd Devices and methods for treating edema
US11717652B2 (en) 2019-02-26 2023-08-08 White Swell Medical Ltd Devices and methods for treating edema
WO2021016372A1 (en) 2019-07-22 2021-01-28 Shifamed Holdings, Llc Intravascular blood pumps with struts and methods of use and manufacture
EP4034192A4 (en) 2019-09-25 2023-11-29 Shifamed Holdings, LLC Intravascular blood pump systems and methods of use and control thereof
US20220125381A1 (en) * 2020-10-23 2022-04-28 Boston Scientific Scimed, Inc. Medical devices and methods for monitoring and treatment with synthetic polymers exhibiting specific binding

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US28742A (en) * 1860-06-19 Machine foe
US486774A (en) * 1892-11-22 William e
US2642874A (en) * 1951-06-04 1953-06-23 Wilmer B Keeling Instrument for treating prostate glands
US3045677A (en) * 1960-05-03 1962-07-24 American Cystoscope Makers Inc Inflatable balloon catheter
US3411506A (en) * 1966-04-11 1968-11-19 Velasco Adolfo Guevara Method and apparatus for hemostasis
US3516408A (en) * 1967-09-21 1970-06-23 Vincent L Montanti Arterial bypass
USRE28742E (en) * 1967-10-26 1976-03-23 Pumps capable of use as heart pumps
US3957389A (en) * 1967-10-26 1976-05-18 Bio-Medicus, Inc. Pumping apparatus and process characterized by gentle operation
US3970408A (en) * 1967-10-26 1976-07-20 Bio-Medicus, Inc. Apparatus for use with delicate fluids
US4037984A (en) * 1967-10-26 1977-07-26 Bio-Medicus, Inc. Pumping apparatus and process characterized by gentle operation
US3647324A (en) * 1969-12-18 1972-03-07 Edson Howard Rafferty Electrically driven pumps capable of use as heart pumps
US3864055A (en) * 1971-12-06 1975-02-04 Harold D Kletschka Pumps capable of use as heart pumps and blood pumps
US3837347A (en) * 1972-04-20 1974-09-24 Electro Catheter Corp Inflatable balloon-type pacing probe
US3995617A (en) * 1972-05-31 1976-12-07 Watkins David H Heart assist method and catheter
US3811800A (en) * 1972-07-12 1974-05-21 K Shill Blood pump
US3851649A (en) * 1973-03-21 1974-12-03 Kendall & Co Catheterization package
SU511951A1 (en) * 1973-07-26 1976-04-30 Всесоюзный научно-исследовательский и испытательный институт медицинской техники Probe for vascular blocking
US4047526A (en) * 1975-05-22 1977-09-13 Sorenson Research Co., Inc. Autologous blood system and method
SU651817A1 (en) * 1976-03-03 1979-03-15 Владивостокский государственный медицинский институт Catheter
US4048064A (en) * 1976-04-23 1977-09-13 Clark Iii William T Biocompatible hemoperfusion system
DE2636290A1 (en) * 1976-08-12 1978-02-16 Fresenius Chem Pharm Ind DEVICE FOR CONTROLLING AND MONITORING BLOOD FLOW DURING BLOOD DIALYSIS, PERFUSION AND DIAFILTRATION USING ONLY ONE CONNECTION POINT TO THE PATIENT'S BLOOD CIRCUIT (SINGLE NEEDLE TECHNOLOGY)
US4300550A (en) * 1978-04-26 1981-11-17 Becton, Dickinson And Company Suction and oxygenation catheter
DE2834956A1 (en) * 1978-08-10 1980-02-21 Thomas Dr Med Urban Double bulb catheter with blood vessel by-pass - for draining blood or administering drugs, comprises two bulbs, tube, and control bulb
US4192302A (en) * 1978-09-12 1980-03-11 Boddie Arthur W Hepatic isolation and perfusion circuit assembly
DE3010841A1 (en) * 1980-03-21 1981-10-08 Ulrich Dr.med. 6936 Haag Uthmann CATHEDER
US4416280A (en) * 1980-04-07 1983-11-22 Minnesota Mining And Manufacturing Company Cardioplegia delivery system
DE3235974A1 (en) * 1981-11-24 1983-06-01 Volkmar Dipl.-Ing. Merkel (FH), 8520 Erlangen DEVICE FOR REMOVAL OR FOR THE EXPANSION OF CONSTRAINTS IN BODY LIQUID LEADING VESSELS
US4423725A (en) * 1982-03-31 1984-01-03 Baran Ostap E Multiple surgical cuff
US4636195A (en) * 1982-04-02 1987-01-13 Harvey Wolinsky Method and apparatus for removing arterial constriction
DE3214397C2 (en) * 1982-04-20 1984-07-26 Karl Dr. 6301 Pohlheim Aigner Double lumen perfusion catheter
US4445892A (en) * 1982-05-06 1984-05-01 Laserscope, Inc. Dual balloon catheter device
DE3228438C2 (en) * 1982-07-30 1985-01-17 Karl Dr. 6301 Pohlheim Aigner Double lumen catheter
DE3309788C2 (en) * 1983-03-18 1985-12-05 Karl Dr. 6301 Pohlheim Aigner Implantable injection catheter
US4883459A (en) * 1983-07-29 1989-11-28 Reynaldo Calderon Retrograde perfusion
US4546759A (en) * 1983-07-29 1985-10-15 Mladen Solar Method and apparatus for assisting human heart function
US4714460A (en) * 1983-07-29 1987-12-22 Reynaldo Calderon Methods and systems for retrograde perfusion in the body for curing it of the disease or immume deficiency
DE3400874C1 (en) * 1984-01-12 1985-02-07 Karl Dr. 6301 Pohlheim Aigner Double-lumen catheter for a device for in-vivo blood purification
US5033998A (en) * 1984-01-20 1991-07-23 Eliot Corday Retrograde delivery of pharmacologic and diagnostic agents via venous circulation
DE3446033A1 (en) * 1984-12-18 1986-07-03 B. Braun Melsungen Ag, 3508 Melsungen IMPLANTABLE INTRAPERITONEAL CATHETER
US4741328A (en) * 1985-03-14 1988-05-03 Shlomo Gabbay Means for intraaortic assist and method of positioning a catheter therefor
US4708718A (en) * 1985-07-02 1987-11-24 Target Therapeutics Hyperthermic treatment of tumors
US4696668A (en) * 1985-07-17 1987-09-29 Wilcox Gilbert M Double balloon nasobiliary occlusion catheter for treating gallstones and method of using the same
US4781677A (en) * 1985-07-17 1988-11-01 Wilcox Gilbert M Method of treatment utilizing a double balloon nasobiliary occlusion catheter
DE3539022C1 (en) * 1985-11-02 1986-10-23 Karl Dr. 6301 Pohlheim Aigner Implantable catheter with vascular throttle
US4705502A (en) * 1985-11-06 1987-11-10 The Kendall Company Suprapubic catheter with dual balloons
US4867742A (en) * 1986-06-06 1989-09-19 Reynaldo Calderon Retrograde perfusion
IT8629545V0 (en) * 1986-06-12 1986-06-12 Fina Ernesto SET BALLOON URETERAL CATHETER BALLOON FOR EXTRACTION OF URETERAL STONES
US4731055A (en) * 1986-08-25 1988-03-15 Becton, Dickinson And Company Blood flow conduit
US4744366A (en) * 1986-09-10 1988-05-17 Jang G David Concentric independently inflatable/deflatable multiple diameter balloon angioplasty catheter systems and method of use
DE3705637A1 (en) * 1987-02-21 1988-09-29 Bissendorf Peptide Gmbh DEVICE FOR REMOVING LOCALLY APPLIED ACTIVE SUBSTANCES AGAINST SOLID TUMORS
US4955857A (en) * 1988-07-18 1990-09-11 Shettigar Udipi R Multi-enzyme bioreactor therapy for cancer
US4913701A (en) * 1988-10-06 1990-04-03 Numed, Inc. Balloon catheter and method of manufacturing the same
US5002559A (en) * 1989-11-30 1991-03-26 Numed PTCA catheter
US5163910A (en) * 1990-04-10 1992-11-17 Mayo Foundation For Medical Education And Research Intracatheter perfusion pump apparatus and method
CA2022019C (en) * 1990-07-26 1992-12-29 Michael Black Catheter
US5161773A (en) * 1990-08-01 1992-11-10 Numed, Inc. Method and apparatus for controlling fluid flow
US5087247A (en) * 1990-08-28 1992-02-11 Cardiovascular Designs, Inc. Balloon perfusion catheter
US5161547A (en) * 1990-11-28 1992-11-10 Numed, Inc. Method of forming an intravascular radially expandable stent
US5188595A (en) * 1991-06-28 1993-02-23 Laserscope Method for enhanced retention of balloon catheter in body cavity

Also Published As

Publication number Publication date
EP0364799A3 (en) 1990-12-27
US5069662A (en) 1991-12-03
EP0364799B1 (en) 1995-05-03
JPH02193676A (en) 1990-07-31
US5411479A (en) 1995-05-02
JP2831056B2 (en) 1998-12-02
EP0364799A2 (en) 1990-04-25
ATE121952T1 (en) 1995-05-15
DE68922456D1 (en) 1995-06-08
DE68922456T2 (en) 1996-01-25
ES2075022T3 (en) 1995-10-01

Similar Documents

Publication Publication Date Title
CA1333872C (en) Cancer treatment
US5919163A (en) Catheter with slidable balloon
US5817046A (en) Apparatus and method for isolated pelvic perfusion
US20010041862A1 (en) Novel apparatus and method of treating a tumor in the extremity of a patient
EP0363886B1 (en) Blood perfusion system and tube means used therein
US6475207B1 (en) Retractable catheter systems and associated methods
US6190371B1 (en) Catheter system having retractable working catheter and associated method
US20120232457A1 (en) Systems and Methods for Homeostatically Treating Organ Disease Using Local Delivery of Therapeutic Agents
US6186146B1 (en) Cancer treatment method
EP0691866B1 (en) Balloon catheter and device for perfusion with the balloon catheter
US6585705B1 (en) Retractable catheter systems
US6905481B2 (en) Percutaneous catheter assembly
US7615034B2 (en) Multi-lumen catheter with protective sheath
JPH10502828A (en) Multilumen catheter system
WO1996034647A1 (en) Implantable catheter and method of use
Canaud et al. Permanent twin catheter: a vascular access option of choice for haemodialysis in elderly patients.
US7022097B2 (en) Method for treating glandular diseases and malignancies
US7914478B2 (en) Intrapelvic perfusion therapy with cancer therapeutic agent and apparatus for intrapelvic perfusion with cancer therapeutic agent
WO2018230577A1 (en) Perfusion device and method for operating same
JP2003102829A (en) Medical method and instrument for perfusing carcinostatic in pelvis
Johnson Complications of vascular access devices
US20050015063A1 (en) Quasi-T shaped catheter for chemotherapy

Legal Events

Date Code Title Description
MKEX Expiry