BIOLOGIC RESPONSE MODIFIERS IN PEDRIATIC CANCER

BIOLOGIC RESPONSE MODIFIERS IN PEDRIATIC CANCER

PEDIATRIC ONCOLOGY IN THE 21st CENTURY, PART I 0889-8588/01 $15.00 + .OO BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER Laura L. Worth, MD, PhD, S...

1MB Sizes 1 Downloads 51 Views

PEDIATRIC ONCOLOGY IN THE 21st CENTURY, PART I

0889-8588/01 $15.00

+ .OO

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER Laura L. Worth, MD, PhD, Sima S. Jeha, MD, and Eugenie S. Kleinerman, MD

Many advances have been made in the development of chemotherapeutic agents, surgical approaches, and radiotherapy for cancer. Despite these advances, several tumors rapidly become resistant to chemotherapy, other tumors grow too close to critical structures to allow their complete surgical removal, and the administration of radiotherapy can be limited by the number of tumor sites involved or the tolerance of the normal tissues in the radiation field. Consequently, new therapeutic approaches are being explored, including the use of biologic response modifiers. The goal of biologic response modifiers is to stimulate the body’s own immune system to help eradicate tumor cells. This stimulation is important because many tumor cells have low or absent major histocompatibility complex (MHC) class I or class I1 molecules, which allows them to escape from immune s~rveillance.~, 60, 88 The use of immunotherapy in the management of cancer began in 1884,12 with the observation of a ”spontaneous regression” of a tumor at the site of an infection. A round cell sarcoma was noted to recur repeatedly after every ”complete” surgical resection. After one surgical excision, an erysipelas infection at the surgical resection site occurred, and the tumor did not recur. This infection was thought to have stimulated the body’s immune system to

Supported in part by CA-42992 (ESK), CA82606 (ESK), and CA16672 Cancer Center Support Core Grant (to The University of Texas M. D. Anderson Cancer Center).

From the Departments of Pediatrics (LLW, SSJ, ESK) and Cancer Biology (ESK), The University of Texas M. D. Anderson Cancer Center, Houston, Texas

HEMATOLOGY/ONCOLOGY CLINICS OF NORTH AMERICA

-

VOLUME 15 NUMBER 4 * AUGUST 2001

723

724

WORTH et a1

help eradicate the residual disease. Based on this observation, Cooley12 directly inoculated an infectious agent to a tumor site in hopes of stimulating an immune response and eradicating the tumor. Immunotherapy has evolved considerably since these early days. The use of socalled biologic response modifiers has become an attractive alternative. Some biologic response modifiers, such as cytokines, are considered natural because the human body produces them. Many biologic response modifiers are less toxic than standard chemotherapeutic agents and result in less damage to the kidneys and liver. Because of their growing importance in the treatment of cancer, biologic response modifiers are now considered the fourth treatment modality, after chemotherapy, radiotherapy, and surgery. This article is divided into three main sections. The first section addresses some of the unique aspects of developing and assessing the effects of biologic response modifiers for clinical trials. The second section looks at biologic response modifiers used in pediatric solid tumors. The final section describes how biologic response modifiers are being used in the treatment of pediatric leukemias and lymphomas. DEVELOPMENT OF BIOLOGIC RESPONSE MODIFIERS

The development of biologic response modifiers differs from that of standard cytotoxic agents. The standard phase I trials used to assess the value of novel chemotherapeutic agents are intended to establish the dose-limiting toxicities of the new agent. Dose-limiting toxicities include bone marrow suppression; breakdown of mucosal membranes; and renal, pulmonary, cardiac, and hepatic toxicities. Because the optimal therapeutic dose for biologic response modifiers is well below the maximally tolerated dose (these agents have a broad dose-response curve), the use of classic phase I trials is not suitable for these compounds. Rather, phase I trials must be designed to measure the specific drug-induced biologic responses created by biologic response modifiers so that the optimal dose for further clinical trials can be determined. Phase I1 or efficacy trials of biologic response modifiers pose special considerations. Many patients who are enrolled on phase I or I1 studies have recurrent or progressive disease that is no longer responsive to chemotherapy. Frequently, these patients have large, bulky disease. Although interleukin (1L)-2 has shown efficacy in advanced renal cell carcinoma and melanoma with bulky disease,I5 many biologic response modifiers are most effective against minimal disease. A biologic response modifier that may be effective against a specific tumor type in the setting of minimal residual disease could end up being discarded in a classic phase I1 trial because it appears to be without activity in the setting of large bulky disease. Biologic response modifiers have mechanisms of action that differ from that of standard chemotherapeutic agents. Frequently, these compounds may take longer to induce an apparent effect. This scenario is illustrated best by the treatment of hemangiomas with

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

725

interferon (IFN)-~L.~~* loo IFN-a acts as an antiangiogenesis factor preventing new vasculature formation. It has little effect on established vasculature. As a result, months of therapy are required before a response is seen. This agent might have been discarded in the classic phase I1 setting if no significant measurable response was noted after only a few cycles of therapy. The definition of a response can differ depending on the intended effect of the biologic response modifier. Some biologic response modifiers are cytostatic, and stable disease, not tumor regression, is the expected outcome. Other biologic response modifiers stimulate an immune response by recruiting lymphocytes and macrophages to the disease site. Such an effect might result in an increased lesion on imaging studies, rather than a decreased one. Consequently, a pulmonary metastasis on standard chest radiography may appear larger rather than smaller after the administration of a biologic response modifier because of an inflammatory response associated with its administration. This change may be mistaken for progressive disease. Additional studies or histologic examination may be needed to show that the enlargement of the mass is not disease progression but rather a fluid-filled cyst or mass devoid of active tumor with an active inflammatory component.10' Many adult tumors respond to the administration of biologic response modifiers. IFN-a alone or in combination with chemotherapeutic agents has been found to be effective in hairy cell leukemia,s3 melan0ma,4~,50, 68 renal cell carcinoma," chronic myeloid leukemia (CML)?2 and multiple myeloma.70,72 IL-2 is an effective agent against renal cell carcinoma and melanoma. Despite documented stimulation of the immune system, however, IL-2 produces no antitumor effect in pediatric sarcomas or neur~blastoma.~ The identification and investigation of agents active specifically in pediatric tumors is a rapidly growing field. BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC SOLID TUMORS

Osteosarcoma

Osteosarcoma is a primary bone tumor that usually presents in adolescents and young adults when the long bones are undergoing rapid growth. Osteosarcoma is an active area of research for biologic response modifiers. Most patients with osteosarcoma have micrometastases at the time of diagnosis.19 Despite surgical resection and aggressive adjuvant chemotherapy, 30% to 40% of patients develop pulmonary metastases. Many of these patients relapse during the first year, usually while still receiving chemotherapy. This finding implies that the metastases are inherently drug-resistant or that the concentration of drug in the lungs is insufficient. Salvage chemotherapeutic regimens have had limited efficacy.41Some of the biologic response modifiers currently being investigated in osteosarcoma are described subsequently (Table 1).

726

WORTH et al

Table 1. BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC SOLID TUMORS Agent

L-MTP-PE ImmTher IL-la IL-2 IL-11 IL-12 IFN-?/ 13-cis-retinoic acid Epogen G-CSF GM-CSF

Proposed Mechanism

Macrophage/ monocyte activator Activation of cytotoxic T lymphocytes Macrophage/monocyte activator Activation of CTL Antiangiogenesis (?) Induction of IL-2, IL-6, colonystimulating factor, tumor necrosis factor, activation of CTLs Stimulate CTL, stimulate the activation and proliferation of NK cells Stimulate platelet production Stimulate IFN-y production, activate NK cell cytotoxicity, activation CTLs Activation of macrophages, NK cells, and CTLs Increase MHC I antigens Differentiation agent Stimulate red blood cell production Stimulate granulocyte progenitor cells Stimulate granulocyte and myeloid progenitor cells

Pediatric Tumor TvDe

Osteosarcoma" Ewing's sarcoma* Osteosarcomat Melanomat Osteosarcoma*

NA Osteosarcomat

Neuroblastoma" NA NA NA

*Clinical trial. tl'reclinical trial. CTL = Cytotoxic T lymphocytes; MHC = major histocompatibility complex; NK = natural killer; G-CSF = granulocyte colony-stimulating factor; GM-CSF = granulocyte-macrophage colonystimulating factor; NA = not appliciable.

Liposome-Encapsulated Muramyl Tripeptide Phosphatidylethanolamine

Muramyl tripeptide phosphatidylethanolamine (MTP-PE) is a derivative of muramyl dipeptide (MDP), the minimal subunit of mycobacteria that is able to activate the immune MTP-PE is lipophilic and can be encapsulated into liposomes (L-MTP-PE).The antitumor effect of MTP-PE is linked to its ability to activate macrophage or monocyte tumoricidal function.28The maximal tolerated dose (MTD) in a standard phase I study was determined to be 6.0 mg/m2, with toxicities limited to flulike symptoms, including chills, fever, myalgias, and fatigue.74 Monocyte tumoricidal activity, the desired biologic response, was maximal at doses of 0.55 to 2.0 mg/m2, however. The monocyte tumoricidal activity as measured by the ability of monocytes isolated from peripheral blood of patients being treated with L-MTP-PE to kill osteosarcoma cells in vitro decreased at the MTD of 6.0 mg/m2, suggesting a suboptimal activation of monocytes at the MTD. Analysis of the serum showed that a dose of 2.0 mg/m2 produced the largest increases in acute-phase reactants, cytokines, white blood cell counts, absolute neutrophil counts, and other indices of stimulated immune function. The dose chosen for the phase I1 and phase I11 study was 2.0 mg/m2, well below the MTD of 6.0 mg/m2.

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

727

Phase I1 trials were crucial in the development of L-MTP-PE. Patients with pulmonary metastases were rendered disease-free by surgery, then treated with L-MTP-PE, the study endpoint being time to relapse. Five patients in this study had a single pulmonary nodule that recurred within 6 weeks of the completion of therapy with L-MTP-PE. Histopathologic examination of these nodules revealed unique features.53Although metastases isolated from patients treated with standard chemotherapeutic agents show central necrosis with a rim of viable tumor at the periphery of a metastasis, lung metastases removed from patients who had received L-MTP-PE showed necrosis and fibrosis at the periphery of the tumor, with viable tumor cells at the center of the lesion. Such tumors are infiltrated with chronic inflammatory cells. This pattern suggested the appearance of pulmonary tuberculosis, in which the infectious lesions are walled off, and necrosis proceeds slowly from the outside so that the lesion is replaced by fibrous t i s s ~ e .This ' ~ finding in lung metastases suggested that the duration of treatment may be crucial for optimal activity of the drug. A significant increase in metastasis-free survival rate was noted for patients treated for 6 months but not for patients who received only 3 months of therapy.52 L-MTP-PE was developed for use with adjuvant chemotherapy and not as a single agent. The antitumor activity of L-MTP-PE is linked directly to its ability to activate the immune function,28,30 whereas chemotherapy in some instances suppresses the immune function. A phase IIb trial designed to assess the interaction between L-MTP-PE and the chemotherapeutic agent ifosfamide showed that ifosfamide did not interfere with the immune system-activating function of L-MTP-PE.51Activation of the immune system by L-MTP-PE did not affect the tumor cytotoxicity of ifosfamide. A randomized phase I11 trial in newly diagnosed osteosarcoma was conducted by the Children's Cancer Group and the Pediatric Oncology Group. The goal of the study was to determine if the addition of ifosfamide, L-MTP-PE, or both to the standard three-drug regimen of doxorubicin, methotrexate, and cisplatin improved the metastasis-free survival rates. Patients were randomized at study entry to one of four different regimens: doxorubicin, methotrexate, and cisplatin with or without L-MTP-PE and doxorubicin, methotrexate, cisplatin, and ifosfamide with or without L-MTP-PE. Although the data are maturing, the four-drug chemotherapy combination with L-MTP-PE (doxorubicin, methotrexate, cisplatin, ifosfamide, and L-MTP-PE) appears to be superior, yielding a disease-free survival rate of 79% at 3 years, compared with 59% seen in patients receiving the four-drug chemotherapy without L-MTP-PE. This improvement in disease-free survival rate is the first to be shown for osteosarcoma in a multicenter study in more than 20 years. Interleukin-la

Other immune modulators that have been used to treat relapsed osteosarcoma include IL-la. This cytokine has many effector functions,

728

WORTH et a1

including the induction of IL-2, IL-6, colony-stimulating factors, tumor necrosis factor (TNF), and intracellular adhesion molecules.6,58, 76 IL-la has been shown to activate T and B cells16,26 and to enhance the cytotoxic activity of lymphocyte^.^^ Used alone, etoposide is not effective against In vitro, however, IL-la increased the cytotoxicity of o~teosarcoma.~~ etoposide against osteosarcoma cells from 9% to 709'0.~~ In a phase I1 study of recombinant IL-la and etoposide in patients with relapsed osteosarcoma, patients were treated daily for 5 days with IL-la, 0.1 mg/ kg, immediately followed by etoposide, 100 mg/m2.101This cycle was repeated every 3 weeks. The study had to be closed because of the unavailability of IL-la. Of the 8 evaluable patients, 3 had stable disease, and 1 had a partial response. The authors of the study concluded that this drug combination warrants further evaluation in a cancer for which few therapeutic options are available for patients with recurrent disease. Future Directions in Osteosarcoma

Because of the success of biologic response modifiers in the treatment of osteosarcoma, the cytokine IL-12 is being investigated. IL-12 exerts a variety of biologic effects on natural killer (NK) and T cells. It activates the cytotoxic activity of NK cells54and facilitates the induction of cytotoxic T lymphocyte^.^^ IL-12 also induces the induction of the Thl response from naive T ~ells,4~, 64 stimulates the production of interferony by NK and T cells, and has been shown to have antiangiogenic activity.94Many of these activities are implicated in the antimetastatic and antitumor effect of IL-12. IL-12 has been shown to be effective when given subcutaneously to patients with renal cell carcinoma73and m e l a n ~ m aIL-12 . ~ administered intravenously in a phase I study produced severe toxic side Because the amount of IL-12 at the tumor site determines the degree of tumor regression, and the main site of relapse in osteosarcoma is the lung, delivery of IL-12 directly to the lung would not only target the site of relapse, but also decrease the side effects associated with systemic therapy. This approach has been investigated in a mouse osteosarcoma model. Mice with microscopic pulmonary metastases were treated with inhalation therapy using an adenoviral vector containing the gene for IL-12. This therapy resulted in the local production of IL-12 in the lung and a dramatic decrease in the number of pulmonary metastases.lo3This approach may prove promising in the treatment of patients with osteosarcoma that has metastasized to the lung. Neuroblastoma

Neuroblastoma, a tumor that arises from neural crest cells, is the most common extracranial solid tumor in children, with approximately 550 new cases each year in the United States. The median age at diagnosis is approximately 22 months. One third of patients with neuro-

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

729

blastoma are less than 1 year old at diagnosis. These patients in general have less advanced disease and a better prognosis. Most patients diagnosed at an age greater than 1 year have tumors that are unresectable or disease that has metastasized to the bone or bone marrow. Despite aggressive therapy, including myeloablative chemotherapy and autologous marrow transplantation, many patients relapse.66 Neuroblastoma is an unusual tumor in that occasional spontaneous regressions have been seen.38,89 If a biopsy specimen is taken from the site where the tumor regressed, differentiated forms of the neuroblastoma, ganglioneuroma, or ganglioneuroblastoma cells are found. This finding suggests that neuroblastoma arises from neuronal crest cells that for some reason do not terminally differentiate. Consequently, drugs that could cause terminal differentiation of these cells could be effective in the treatment of this disease.

134s -Retinoic Acid In vitro, 13-cis-retinoic acid is a vitamin A derivative shown to decrease the proliferation and to cause the differentiation of neuroblastoma cell lines even when the cells are resistant to cytotoxic therapy.69,85 A study was developed to determine if 13-cis-retinoic acid could have the same effect on neuroblastoma cells in vivo as it did in vitro. In a prospective randomized trial, patients received standard chemotherapy with or without 13-cis-retinoic acid or autologous bone marrow transplant with or without 13-cis-retinoic acid. Although the highest diseasefree survival rate was seen in the group that received 13-cis-retinoic acid after autologous bone marrow transplant, the survival rate of patients who also received 13-cis-retinoic acid and standard chemotherapy was higher than that of patients who received chemotherapy alone.66These results suggest that 13-cis-retinoic acid may be inducing the differentiation of neuroblastoma in the presence of minimal residual disease. More information on the use of retinoids in the management of childhood cancer is given elsewhere in this issue. Monoclonal Antibodies Monoclonal antibodies (mAbs) can be used for diagnosis, identification of metastases, and therapy. In pediatric cancers, a-fetoprotein (hepatic tumors), human chorionic gonadotropin (germ cell tumors), and GD2 (neuroblastoma) are tumor-associated antigens that aid in the diagnosis and tracking of tumor progression. GD2 is a disialoganglioside expressed on tumors of neuroectoderma1 origin. GD2 also is expressed in the cerebellum and on peripheral nerves. Tumor imaging with I3l3F8, a murine mAb directed toward GD2, has aided in the detection of metastatic disease and in planning for radiotherapy for neuroblastoma. This radiolabeled antibody can be used to deliver radiation. The use of this antibody to deliver doses of 4000 cGy to the tumor has resulted in a survival advantage for patients with

ne~roblastoma.~~ The anti-GD2 antibody, 14.G2a, facilitates antibodydependent cytotoxicity. In a phase I trial of 14.G2a in combination with IL-2, responses were noted in patients with neuroblastoma, most of whom had bulky disease. Twenty-six patients entered in this trial completed a full course of therapy and were evaluable. Of the 24 evaluable patients with neuroblastoma, 7 had stable disease, and 1 had a partial response. Of the 10 evaluable neuroblastoma patients with marrow disease, 3 had a significant decrease in the percentage of neuroblastoma cells in the bone marrow.34Further investigation of the combination of 14.G2a and IL-2 will involve its use in the setting of minimal residual disease or in an adjuvant setting. Ewing’s Sarcoma

Ewing’s sarcoma is part of a family of tumors that range from sarcoma of the bone and soft tissue to more mature cell types that express neuronal differentiation as found in the peripheral primitive neuroectodermal tumors. Using multimodality treatment plans, advances have been made in the management of Ewing’s sarcoma. Patients with primary tumors of the humerus, femur, or trunk; patients with metastases at diagnosis; and patients with bulky disease have a high risk of relapse, however.44,99 Although high-dose, short-term therapy with vincristine, doxorubicin, and cyclophosphamide (VAC therapy) increased the initial complete response rate in patients with high-risk Ewing’s sarcoma, the relapse rate, 40% to 50% at 2 years, remained high.56Relapsed Ewing’s sarcoma is particularly difficult to treat because of drug resistance and because the incidence of second remissions is extremely low. Two immune-modulating approaches are being investigated: one using a macrophage activator and one using cytokines in the setting of isolated limb perfusion. Liposome-Encapsulated Disaccharide Tripeptide (ImmTher)

Regardless of the sensitivity to chemotherapy, most Ewing‘s sarcoma tumor cells can be killed by activated macro phage^.^^, lo2 The efficacy of dose-intensive VAC therapy in combination with the macrophage-activating agent ImmTher was in~estigated.~~ ImmTher is a new liposome-encapsulated lipophilic disaccharide tripeptide derivative of MDP. The liposomal compound is similar to MTP-PE in that it activates monocyte tumoricidal activity and stimulates the production of IL-1, IL6, IL-8, and TNF. In vitro, however, the disaccharide tripeptide moiety ImmTher was more tumoricidal than MDP. In contrast to MTP-PE, ImmTher increases the expression and production of IL-12 by monocytes,lo3which may enhance the antitumor activity of the drug. ImmTher stimulates human monocytes in vitro to kill several different human Ewing’s sarcoma cell lines.lo3ImmTher ’s role against pulmonary metas-

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

731

tases in Ewing’s sarcoma is being investigated in a weekly schedule given for 52 weeks after the completion of dose-intensive VAC chemotherapy, surgery, radiotherapy, or a combination of these modalities. Tumor Necrosis Factor and Interferony in Limb Perfusion

Another innovative approach to treating extremity soft tissue sarcomas is isolated limb perfusion for the delivery of chemotherapeutic agents and cytokines. This approach was developed by Eggermont et a1,2O who used isolated limb perfusion under hyperthermic conditions with high-dose TNF-a, IFN-y, and melphalan. Because of the systemic side effects of IFN-y, this trial was repeated using only TNF-a and melphalan. The response rate of 81°/P was similar to that obtained using all three agents.20Because of the success in delivering biologic response modifiers by isolated limb perfusion, this approach is being investigated as a gene delivery Malignant Brain Tumors

Tumors of the central nervous system are the most common malignant solid tumor in children and are challenging to cure. Changes in chemotherapy have resulted in only limited progress in central nervous system tumors, and the prognosis after frontline therapy has failed is dismal. The role of biologic response modifiers is being investigated in this setting. Interferon-p

One agent being used in brain tumors is IFN-P. Results have been mixed. A study in Japan reported the effectiveness of IFN-P in combination with carmustine and radiotherapy in 10 of 11patients.96In 2 patients with recurrent primitive neuroectodermal tumors, IFN-P given daily as a single agent resulted in stable disease, with minimal side effects. At the time of the study, the patients had been treated for 14 and 24 months.93The Children’s Cancer Group conducted a phase 1/11 study of IFN-P and hyperfractionated radiotherapy in children with newly diagnosed brainstem gliomas. The investigators concluded that this combination could be tolerated by this group of patients, with occasional dose-limiting toxicities of the liver, blood, and central nervous system. This therapy did not improve the rate of disease control, however, and 30 of 32 patients developed progressive disease at a median of 5 months and died at a median of 9 months after study entry.*l Interleukin-13

IL-13 is an immune regulatory cytokine that is structurally homologous to IL-4. It is unclear why malignant glioma cells have an abundance

732

WORTH et al

of IL-13 receptors on their surface, but they make this receptor an attractive target for antiglioma therapies.14 An antagonist to IL-4, hIL4.Y124D, inhibits the physiologic effect of IL-4 and IL-13 on normal cells. This same antagonist to IL-4 does not inhibit the IL-13-based cytotoxicity on glioma cells, however. This finding suggests that the IL13 receptors on glioma cells are tumor specific in the presence of an IL4 antagonist and that this antagonist can serve as a receptor-directed cytotoxin without being cytotoxic to normal cells. Hematopoietic Growth Factors in Solid Tumors

Hematopoietic growth factors do not play an important role in the modulation of the immune system in pediatric solid tumors. Instead, .these growth factors frequently are used to stimulate the recovery of granulocytes (e.g., granulocyte-colony stimulating factor [G-CSF]) after chemotherapy so that the dose of chemotherapy can be increased or the time between courses of chemotherapy can be shortened.47G-CSF is used to stimulate the production and mobilization of stem cells for use in transplantation. Epogen, which is created using recombinant DNA technology, has the same amino sequence as erythropoietin. Epogen stimulates red blood cell productionz and has been used to decrease the need for red blood cell transfusions in patients receiving chemotherapy. The use of IL-11, a growth factor produced by stromal cells, increases platelet counts.'* Its use in breast cancer patients treated with multiple cycles of cyclophosphamide and doxorubicin reduced the incidence of severe thromb~cytopenia.~~ BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC HEMATOLOGIC MALIGNANCIES

Anecdotal reports of spontaneous remissions of leukemia after blood transfusions suggest that the immune system plays an important role in regulating leukemia cell proliferation. Based on Cooley's observation of the regression of a sarcoma in the face of infection, early attempts using immune modulation involved nonspecific stimulation of the immune system in hopes of stimulating a nonspecific activation of the immune system. These early approaches used bacille Calmette-Gukrin, a modified form of the tubercle bacillus, because it had shown increased immune reactions in murine models. When used in patients with advanced disease, however, these approaches were almost uniformly unsuccessful, and these efforts have largely been abandoned? Data from allogeneic bone marrow transplant recipients showed that the immune system plays a crucial role in eradicating minimal residual leukemia: Tcell depletion decreases the incidence and severity of graft-versus-host disease but is associated with a higher relapse rate.I7 Donor leukocyte transfusion can induce remission in CML patients who relapse after

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

733

allogeneic BMT.82Various studies suggest that a cure does not always entail total eradication of leukemia but might rather be a state in which the immune system keeps minimal residual disease under check.86Advances in molecular biology have allowed a better characterization of the various lineages of hematopoietic cells, making these easily accessible cells ideal targets for biologic therapies. Although the use of cytokines continues to be explored and improved, mAbs are being used in various treatment regimens for leukemia.

Monoclonal Antibodies

The development of techniques for generating mAbs has contributed to the armamentarium of targeted leukemia therapy. After attaching to the cell surface, unmodified mAbs activate cell-mediated and complement-mediated cytotoxicity mechanisms, causing cell death.'O The development of recombinant chimeric mAbs that use the constant region of human antibodies and the variable combining region of the mouse antibody had decreased the incidence of the development of human antimurine antibodies (HAMA), improving the potency of mAbs and allowing repeat infusion^.^, 37 Radioactive isotopes and toxins can be conjugated to mAbs to increase their effectiveness against mAbs can be used to target any leukemia cell surface marker. The efficacy of anti-CD20, anti-CD52, and anti-CD33 mAbs is well established. Researchers are studying the use of these agents to intensify chemotherapy, to consolidate maintenance, and to purge stem cells before transplant. IDEC-C2B8 (IDEC- 102, Rituximab)

The surface antigen CD20 is expressed in more than 90% of cells of B-cell lymphomas and in about 50% of cells of B-cell precursor acute lymphocytic leukemias (ALL). CD20 is not shed by the cells; does not circulate in the plasma as a free protein; and is not internalized, modulated, or down-regulated on antibody binding. IDEC-C2B8 (IDEC-102, rituximab) is a chimeric humanized mAb that binds to CD20.84Used in relapsed and refractory low-grade and follicular non-Hodgkin's lymphoma, rituximab has shown response rates of 48% in several trials.6143 In 83% of the patients, circulating B cells were depleted within the first 3 doses, with sustained depletion for 6 to 9 months. Although rituximab depletes normal B cells, it has not been associated with profound immunosuppression. Of initial responders, 40% benefit from repeat therapy after disease progression. Less than 1%of patients develop quantifiable immunoreactivity (HAMA or human antichimeric antibodies [HACA]). Treatment-related toxicity is limited primarily to infusion-related events, including fever, chills, skin rash, nausea, headache, rigor, hypotension, and bronchospasm. These events usually are associated with the first

infusion, are believed to be due to cytokine release, and can be reversed by adjusting the infusion rate. Although rituximab is licensed for the treatment of indolent lymphoma, the role of this mAb in combination with chemotherapy in CD20-positive lymphomas and leukemias is being investigated. The use of rituximab is being investigated for the treatment of Hodgkin’s disease. Approximately 25% of Hodgkin’s lymphomas are CD-20 positive. Some patients with CD20-negative Hodgkin’s disease have responded to rituximab therapy. Although rituximab did not target specifically the tumor cells in the latter cases, it suppressed the normal B cells that sustain Reed-Sternberg cell proliferation. A trial with rituximab in combination with gemcitabine is currently under way for patients with relapsed Hodgkin’s disease. CAMPATH- 1H

CAMPATH-1H is a humanized mAb specific for the surface antigen CD52, a cell surface glycoprotein that is expressed abundantly on most normal and malignant lymphocytes and monocytes. CAMPATH-1H was used initially to control graft-versus-host disease and graft rejection post transplantation.4°,97 The demonstrable immunosuppressive effects of this agent led to its use in autoimmune diseases.8,59 The drug now is under investigation as part of a nonmyeloablative conditioning regimen before allogeneic stem cell transplantation. Although CAMPATH-1H induced only a modest overall response rate in non-Hodgkin’s lymphoma patients with bulky lymph nodes, significant disease regression was seen in blood and bone marrow.39CAMPATH-1H was shown later to be highly effective and well tolerated in patients with chronic lymphocytic leukemia (CLL).*OThe role of CAMPATH-1H as a therapy for minimal versus active disease or single use versus its use in combination with chemotherapy remains to be established. CMA-676 (Gemtuzumab, Zogamicin, Mylotarg)

The surface antigen CD33 is highly expressed on the myeloid blast cells of most patients with acute myeloid leukemia (AML). Mylotarg is a conjugate of humanized anti-CD33 mouse mAb and calicheamicin, a highly potent antitumor antibiotic that is inactive when bound to the mAb.43After binding to the surface of the AML cell, the immunotoxin conjugate is internalized. Within the cell, the antibody linker is hydrolyzed, liberating free calicheamicin, which diffuses to the nucleus and cleaves double-stranded DNA. In preliminary studies, 30% of patients with relapsed AML responded to treatment with m y l ~ t a r g .91~ ~ , Chills, fever, hypotension, and dyspnea were reported shortly after the completion of infusion in some patients. These symptoms usually are mild and reversible. Myelosuppression, especially neutropenia and thrombocytopenia, is common. Mylotarg would seem to be an effective therapy for AML in first relapse. This agent has a favorable safety profile

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

735

and is the only antibody-targeted chemotherapeutic agent approved for use in the United States for the treatment of AML. Ongoing studies are evaluating different doses and schedules of mylotarg alone and in combination with other therapies. Cytokines

The use of G-CSF and granulocyte-macrophage (GM)-CSF in hematologic malignancies allows for safe and timely administration of intensive therapy by shortening the duration of neutropenia. These agents also are used to mobilize stem cells. The use of G-CSF and GM-CSF as recruiting and synchronizing agents to enhance the cytotoxicity of cytosine arabinoside has failed to show a priming advantage.23,77 The use of either factor is not associated with higher relapse rates, however, and they probably are safe when used in the setting of low tumor burden. IL-2 stimulates the proliferation of T cells and induces the elaboration of other cytokines, including IFN-y and TNF.42IL-2 activates NK cells to become lymphokine-activated killer cells.79IL-2 can block leukemia cell growth in vitro and in mouse models. Initial work focused on the use of IL-2 in the setting of advanced disease.87Clinical trials have shown that IL-2 is most effective against low leukemic burden.31,32, 65, 71 Current studies are focusing on the use of IL-2 in patients in remission and post transplantation.', 98 Preliminary evidence indicates that IL-2 may be active in AML but not in ALL. IFN-a revolutionalized the treatment of CML in the 1 9 8 0 ~IFN-a .~~ has shown sporadic success in inducing remissions in childhood ALL but has never achieved the consistent results observed in hairy cell leukemia or CML. Hematologic response is observed in most CML patients treated with IFN-a, with 26% achieving complete cytogenetic response. A subset of these patients have been followed up without maintenance treatment and have remained in cytogenetic remission for 10 years. Researchers developed a pegylated (PEG)-IFN-a, which is a form of rHu-IFN alfa-2a that has been modified chemically by the covalent attachment of a branched methoxypolyethylene glycol moiety. Data from animals and humans indicate that PEG-IFN-a injected once a week is more effective and tolerated better than IFN-a injected 3 times a week. Combined with the tyrosine kinase inhibitor STI-571, discussed elsewhere in this issue, PEG-IFN-a might provide an effective targeted therapy for CML and avoid the morbidity of bone marrow transplant, which is the current standard of care. CONCLUSIONS

Biologic response modifiers are becoming an important addition to surgery, chemotherapy, and radiotherapy in the management of cancer. As this field of research grows and expands, more biologic response modifiers will be incorporated into therapeutic regimens. By stimulating

736

WORTH et a1

the immune system to eradicate minimal residual disease, these agents may improve the disease-free and long-term survival rates of patients with a variety of malignancies. The challenge is to incorporate biologic response modifiers into the treatment armamentarium in ways that will maximize their tumorigenicity.

References 1. Ackerstein A, Kedar E, Slavin S: Use of recombinant human interleukin-2 in conjunction with syngeneic bone marrow transplantation in mice as a model for control of minimal residual disease in malignant hematologic disorders. Blood 783212, 1991 2. Akporiaye ET, Hersh EM: Immune adjuvants. In DeVita VT, Hellman S, Rosenberg SA (eds): Biologic Therapy of Cancer, ed 2. Philadelphia, Lippincott-Raven, 1995, 635 . 3. Atkins MB, Robertson MJ, Gordon M, et al: Phase I evaluation of intravenous recombinant human IL-12 in patients with advanced malignancies. Clin Cancer Res 3:409, 1997 4. Bajetta E, Del Vecchio M, Mortarini R, et al: Pilot study of subcutaneous human recombinant interleukin-12 in metastatic melanoma. Clin Cancer Res 475, 1998 5. Bauer M, Reaman GH, Hank JA, et al: A phase I1 trial of human recombinant interleukin-2 administered as a 4-day continuous infusion for children with refractory neuroblastoma, non-Hodgkin‘s lymphoma, sarcoma, renal cell carcinoma, and malignant melanoma: A Children’s Cancer Group Study. Cancer 75:2959, 1995 6. Belardelli F, Proietti E, Ciolli V, et al: Interleukin-lp induces tumor necrosis and early morphological and metabolic changes in transplantable mouse tumors: Similarities with the anti-tumor effects of tumor necrosis factor ci or p. Int J Cancer 44:116, 1989 7. Bodmer WF, Browning MJ, Krausa P, et al: Tumor escape from immune response by variation in HLA expression and other mechanisms. Ann N Y Acad Sci 690:42, 1993 8. Calne R, Moffatt SD, Friend PJ, et a1 CAMPATH-1H allows low-dose cyclosporine monotherapy in 31 cadaveric renal allograft recipients. Transplantation 68:1613, 1999 9. Caron PC, Jurcic JG, Scott AM, et al: A phase IB trial of humanized monoclonal antibody M 195 (anti-CD33)in myeloid leukemia: Specific targeting without immunogenicity. Blood 83:1760, 1994 10. Caron PC, Scheinberg DA: Immunotherapy for acute leukemias. Curr Opin Oncol 6:14, 1994 11. Cohen J: IL-12 deaths: Explanation and a puzzle. Science 270:908, 1995 12. Cooley W The treatment of malignant tumors by repeated inoculations of erysipelas: A report of ten original cases. Am J Med Sci 105:487, 1893 13. Dannenberg AM Jr, Thomashetski F Jr: Pathogenesis of pulmonary tuberculosis. In Fisherman AP (ed): Pulmonary Disease and Disorders, ed 2. New York, McGrawHill, 1988, p 1821 14. Debinski W, Gibo DM, Puri RK: Novel way to increase targeting specific to a human glioblastoma-associated receptor for interleukin 13. Int J Cancer 76:547, 1998 15. Dillman R O The clinical experience with interleukin-2 in cancer therapy. Cancer Biother 9:183, 1994 16. Dinarello CA: Biology of interleukin 1. J Exp Med 162:501, 1985 17. Drobyski WR, Keever CA, Roth MS, et al: Salvage immunotherapy using donor leukocyte infusions as treatment for relapsed myelogenous leukemia after allogeneic bone marrow transplantation: Efficacy and toxicity of a defined T-cell dose. Blood 82:2310, 1993 18. Du XX, Williams MA: Interleukin-11: A multifunctional growth factor derived from the hematopoietic microenvironment. Blood 9283, 1995 19. Eilber F, Guiliano A, Eckardt J, et al: Adjuvant chemotherapy for osteosarcoma: A randomized prospective trial. J Clin Oncol 5:21, 1987 20. Eggermont A, Koops H, Lienard D, et al: Isolated limb perfusion with high-dose

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

737

tumor necrosis factor-a in combination with interferon-a and melphalan for nonresectable extremity soft tissue sarcoma: A multicenter trial. J Clin Oncol 142653,1996 21. Eggermont A, Schraffordt Koops H, Klausner JM, et al: Isolated limb perfusion with tumor necrosis factor alpha and chemotherapy for advanced extremity soft tissue sarcoma. Semin Oncol 24:542, 1997 22. Egrie JC, Strickland TW, Lane J, et al: Characterization and biological effects of recombinant human erythropoietin. Immunobiology 72:213, 1986 23. Estey EH: Use of colony-stimulating factors in acute myeloid leukemia. Blood 832015, 1994 24. Ezekowitz RAB, Mulliken JB, Folkman J: Interferon alfa-2a therapy for life-threatening hemangiomas of infancy. N Engl J Med 326:456, 1992 25. Fader1 S, Talpaz M, Estrov Z, et al: Chronic myelogenous leukemia: Biology and therapy. Ann Intern Med 131:207, 1999 26. Farran W, Mizel S, Farran J: Participation of lymphocyte activation factor (IL-I) in the induction of cytotoxic T-cell responses. J Immunol 124:1371, 1980 27. Fidler IJ: Immunomodulation of macrophages for cancer and antiviral therapy. In Thomlinson E, Davis SS (eds): Site-Specific Drug Delivery. New York, Wiley 1986, p 111 28. Fidler IJ, Barnes Z, Fogler WE, et al: Involvement of macrophages in the eradication of established metastases following the intravenous injection of liposomes containing macrophage activators. Cancer Res 42:496, 1982 29. Fidler IJ, Kleinerman ES: Lymphokine-activated human blood monocytes destroy tumor cells but not normal cells under co-cultivation conditions. J Clin Oncol 2:937, 1984 30. Fidler IJ, Nii A, Tsau JY, et al: Activated human blood monocytes trigger the antitumor activity of blood polymorphonuclear cells. Lymphokine Res 8:427, 1989 31. Foa R Does interleukin-2 have a role in the management of acute leukemia. J Clin Oncol 11:1817, 1993 32. Foa R, Meloni G, Tosti S, et al: Treatment of acute myeloid leukaemia patients with recombinant interleukin-2: A pilot study. Br J Haematol 77491, 1992 33. Friesen C, Herr I, Krammer PH, et al: Involvement of the CD95 (APO-l/FAS) receptor-ligand system drug-induced apoptosis in leukemia cells. Nat Med 2:574,1996 34. Frost JD, Hank JA, Reaman GH, et al: A phase I/IB trial of murine monoclonal antiGD2 antibody 14.g2a plus interleukin-2 in children with refractory neuroblastoma: A report from the Children’s Cancer Group. Cancer 80:317, 1997 35. Gately MK, Wolitzky AG, Quinn PM, et al: Regulation of human cytolytic lymphocyte responses by interleukin-12. Cell Immunol 143:127, 1992 36. Gordon MS, Hoffman R, Battiato L, et al: Recombinant human interleukin eleven (Neumega rhIL-11 growth factor; rhIL-11) prevents severe thrombocytopenia in breast cancer patients receiving multiple cycles of cyclophosphamide (C) and doxorubicin (A) chemotherapy [abstr]. Proc ASCO 13:A326, 1994 37. Grossbard ML, Press OW, Appelbaum FR, et al: Monoclonal antibody-based therapies of leukemia and lymphoma. Blood 80:863, 1992 38. Haas D, Ablin AR, Miller C, et a1 Complete pathological maturation and regression of Stage IVS neuroblastoma without treatment. Cancer 62:818, 1988 39. Hale G, Dyer MJ, Clark MR, et al: Remission induction in non-Hodgkin’s lymphoma with reshaped human monoclonal antibody CAMPATH-IH. Lancet 2:1394,1988 40. Hale G, Zhang MJ, Bunjes D, et a 1 Improving the outcome of bone marrow transplantation by using CD52 monoclonal antibodies to prevent graft-versus-host disease and graft rejection. Blood 92:4581, 1998 41. Harris MB, Cantor A, Goorin A, et al: Response to ifosfamide in patients with osteosarcoma: A comparison of results in newly diagnosed patients versus those with recurrent disease after adjuvant chemotherapy [abstr]. Proc ASCO 10:315, 1991 42. Heslop HE, Gottlieb DJ, Reittie JE, et al: Spontaneous and interleukin-2 induced secretion of tumor necrosis factor and gamma interferon following autologous marrow transplantation or chemotherapy. Br J Haematol 72:122, 1989 43. Hinman LM, Hamann PR, Wallace R, et al: Preparation and characterization of

738

WORTH et a1

monoclonal antibody conjugates of the calicheamicins: A novel and potent family of antitumor antibiotics. Cancer Res 53:3336,1993 44. Horowitz ME, Kinsella TJ, Wexlerm LH, et al: Total-body irradiation and autologous bone marrow transplant in the treatment of high-risk Ewing’s sarcoma and rhabdomyosarcoma. J Clin Oncol 11:1911,1993 45. Hsieh CS, Matacatoma SE, Tripp CS, et al: Development of the Thl CD4’ cells through IL-12 produced by Listeria-induced macrophages. Science 260:542, 1993 46. Jia S-F, McWatters A, Kleinerman ES: Synergistic antitumor effect of VP-16 and IL-1, TNF alpha or IFN gamma against human osteosarcoma cells [abstr]. Proc Am Assoc Cancer Res 35:A3116, 1994 47. Johnston EM, Crawford J: Hematopoietic growth factors in the reduction of chemotherapeutic toxicity. Semin Oncol25:552, 1998 48. Jurcic JC, Scheinberg DA. Recent developments in the radioimmunotherapy of cancer. Curr Opin Immunol6:715, 1994 49. Kirkwood JM, Strawderman MH, Ernstoff MS, et al: Interferon-alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: The Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 147, 1996 50. Kirkwood J: Biological therapy with interferon alpha and beta: Clinical applicationsmelanoma. In De Vita VJ, Hellman S, Rosenberg S (eds): Biological Therapy of Cancer: Principles and Practice. Philadelphia, JB Lippincott, p 388, 1995 51. Kleinerman ES, Meyers PA, Raymond AK, et al: Combination therapy with ifosfamide and liposome-encapsulated muramyl tripeptide: Tolerability, toxicity and immune stimulation. J Immunother 17181, 1995 52. Kleinerman ES, Gano JB, Johnston DA, et al: Efficacy of liposomal muramyl tripeptide (CGP19835A) in the treatment of relapsed osteosarcoma. Am J Clin Oncol 18:93,1995 53. Kleinerman ES, Raymond AK, Bucana CD, et al: Unique histological changes in the lung metastases of osteosarcoma patients following therapy with liposomal muramyl tripeptide (CGP 19835A lipid). Cancer Immunol Immunother 34211,1992 54. Kobayashi M, Fitz L, Ryan M, et al: Identification and purification of a natural killer cell stimulatory factor (NKSF), a cytokine with multiple biological effects on human lymphocytes. J Exp Med 170:827, 1979 55. Kung F, Hayes FA, Krischer J, et al: Clinical trial of etoposide (VP-16) in children with recurrent malignant solid tumors: A phase I1 study from the Pediatric Oncology Group. Invest New Drugs 631, 1988 56. Kushner BH, Meyers PA, Gerald WL, et al: Very-high-dose short-term chemotherapy for poor-risk peripheral primitive neuroectodermal tumors, including Ewing’s sarcoma, in children and young adults. J Clin Oncol 132796, 1995 57. Larson SM, Sgouros G, Cheung NK: Antibodies in cancer therapy: Radioisotope conjugates. In DeVita VT, Hellman S, Rosenberg SA (eds): Biological Therapy of Cancer, ed 2. Philadelphia, JB Lippincott, 1995, p 534 58. Le J, Vilcek J: Tumor necrosis factor and interleukin 1: Cytokines with overlapping biological activities. Lab Invest 56934, 1987 59. Lim SH, Hale G, Marcus RE, et al: CAMPATH-1 monoclonal antibody therapy in severe refractory autoimmune thrombocytopenic purpura. Br J Haematol84:542,1993 60. Main EK, Monos DS, Lampson LA: IFN-treated neuroblastoma cell lines remain resistant to T cell-mediated allo-killing and susceptibility to non-MHC restricted cytotoxicity. J Immunol 1412943, 1998 61. Maloney DG, Liles TM, Czerwinski C, et al: Phase I clinical trial using escalating single-dose infusion of chimeric anti-CD20 monoclonal antibody (IDEC-C2B8) in patients with recurrent B-cell lymphoma. Blood 842457, 1994 62. Maloney DG, Grillo-Lopez AJ, Bodkin D, et al: IDEC-C2B8: Results of a phase I multiple-dose trial in patients with relapsed non-Hodgkin’s lymphoma. J Clin Oncol 15:3266, 1997 63. Maloney DG, Grillo-Lopez AJ, White CA, et a1 IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin’s lymphoma. Blood 90:2188, 1997 64. Manetti R, Parronchi P, Guidizi MG, et al: Natural killer cell stimulatory factor

BIOLOGIC RESPONSE MODIFIERS IN PEDIATRIC CANCER

739

interleukin-12 (IL-12) induces T helper (Th1)-specificimmune responses and inhibits the development of IL-4 producing Th cells. J Exp Biol 1771199, 1993 65. Maraninchi D, Blaise D, Viens P, et al: High-dose recombinant interleukin-2 and acute myeloid leukemia in relapse. Blood 78:2182, 1991 66. Matthay KK, Villablanca JG, Sugar RC, et al: Treatment of high-risk neuroblastoma with intensive chemotherapy, radiotherapy, autologous bone marrow transplant, and 13-cis-retinoic acid. N Engl J Med 241:1165, 1999 67. Milas M, Feig B, Yu D, et al: Isolated limb perfusion in the sarcoma-bearing rat: A novel preclinical gene delivery system. Clin Cancer Res 3:2197, 1997 68. McLeod GRC, Thomson DB, Hersey P: Recombinant interferon-alfa 2a in advanced malignant melanoma: A phase 1-11 study in combination with DTIC. Int J Cancer l(suppl):31, 1987 69. Melino G, Thiele C, Knight RA, et al: Retinoids and the control of growth/death decisions in human neuroblastoma cell lines. J Neurooncol31:68, 1997 70. Melldtedt H, Ahre A, Bjorkholm M, et al: Interferon therapy in myelomatosis. Lancet 1:245, 1979 71. Meloni G, Vignetti M, Andrizzi C, et al: Interleukin-2 treatment in acute myeloblastic leukemia in different phases of disease. Blood M2158, 1994 72. Montuoro A, De Rosa L, De Blasio A, et al: Alpha-2a-interferon/melphalan/prednisone versus melphalan/prednisone in previously untreated patients with multiple myeloma. Br J Haematol 76365, 1990 73. Motzer RJ, Rakhat A, Schwartz LH, et al: Phase I trial of subcutaneous recombinant human interleukin-12 in patients with advanced renal cell carcinoma. Clin Cancer Res 41183, 1998 74. Murray JL, Kleinerman ES, Cunningham JE, et al: Phase I trial of liposomal muramyl tripeptide phosphatidylethanolamine in cancer patients. J Clin Oncol 71915, 1989 75. Neidhart J, Anderson S, Harris J, et al: Vinblastine fails to improve response to renal cancer to interferon alfa-nl: High response rate in patients with pulmonary metastases. J Clin Oncol9:832, 1991 76. Ochoa AC, Gromo G, Alter BJ, et al: Long-term growth of lymphokine-activated killer (LAK) cells: Role of anti-CD3, beta-IL 1, and interferon-gamma and-beta. J Immunol 138:2728, 1987 77. Ohno R, Tomonaga M, Kobayashi T, et al: Effect of granulocyte colony-stimulating factor after intense induction therapy in relapsed or refractory acute leukemia. N Engl J Med 323:871, 1990 78. Oppenheim JJ, Kovacs EJ, Matsushima K There is more than one interleukin-1. Immunology 745,1986 79. Oshimi K, Oshimi Y, Akutsu M, et a1 Cytotoxicity of interleukin-2 activated lymphocytes for leukemia and lymphoma cells. Blood 68:938, 1986 80. Osterborg A, Dyer MJ, Bunjes D, et al: Phase I1 multicenter study of human CD52 antibody in previously treated chronic lymphocytic leukemia. European Study Group of CAMPATH-1H Treatment in Chronic Lymphocytic Leukemia. J Clin Oncol 15:1567, 1997 81. Packer RJ, Prados M, Phillips P, et al: Treatment of children with newly diagnosed brain stem gliomas with intravenous recombinant beta-interferon and hyperfractionated radiation-therapy: A Children’s Cancer Group phase 1/11 study. Cancer 772150, 1996 82. Porter DL, Roth MS, McGarigle C, et al: Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia. N Engl J Med 330:100, 1994 83. Quesada JR, Rueben J, Manning JT, et al: Alpha interferon for induction of remission in hairy cell leukemia. N Engl J Med 310:15, 1984 84. Reff ME, Camer C, Chambers KS, et al: Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 83:435, 1994 85. Reynolds CD, Kane DI, Einhorm PA, et al: Response of neuroblastoma to retinoic acid in vitro and in vivo. Prog Clin Biol Res 366:203, 1991 86. Roberts WM, Estrov Z, Ouspenskaia M, et al: Measurement of residual leukemia during remission in childhood acute lymphoblastic leukemia. N Engl J Med 336:317, 1997

740

WORTH et a1

87. Rosenberg SA, Lotze MT, Muul LM, et al: A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin alone. N Engl J Med 316:889, 1987 88. Roth C, Rochlitz C, Kourilsky I? Immune response against tumors. Adv Immunol 57:281, 1994 89. Schwartz AD, Dadash-Zadeh M, Lee H, et al: Spontaneous regression of disseminated neuroblastoma. J Pediatrics 85:760, 1974 90. Sievers EL, Appelbaum FA, Spielberger RT, et a1 Selective ablation of acute myeloid leukemia using antibody-targeted chemotherapy: A phase I study of an anti-CD33 calicheamicin immunoconjugate. Blood 93:3678, 1999 91. Sievers EL, Larson RA, Estey E, et al: Preliminary results of the efficacy and safety of CMA-676 in patients with AML in first relapse. Proc ASCO 18:7a, 1999 92. Talpaz M, Kantarjian HM, McCrediek, et al: Hematological remission and cytogenetic improvement induced by recombinant human interferon alpha A in chronic myelogenous leukemia. N Engl J Med 314:1065,1986 93. Tanaka S, Okada M, Murakami M, et a1 Human interferon-beta therapy for cerebral primitive neuroectodermal tumor-two case reports. Neurol Med Chir (Tokyo) 35:82, 1995 94. Voest EE, Kenyon BM, OReilly MS, et al: Inhibition of angiogenesis is vivo by interleukin 12. J Natl Cancer Inst 87571, 1996 95. Vosika GJ, Cornelius DA, Bennek SA, et a1 Immunological and toxicologic study of disaccharide tripeptide glycerol dipalmitoyl: A new lipophilic immunomodulator. Mol Biother 250, 1990 96. Wakabayashi T, Yoshida J, Mizuno M, et al: Effectiveness of interferon-beta, ACNU, and radiation therapy in children with brainstem gliomas. Neurol Med Chir (Tokyo) 32:942, 1992 97. Waldmann H, Polliak A, Hale G, et al: Elimination of graft-versus-host-disease by in vitro depletion of alloreactive lymphocytes with a monoclonal rat anti-human lymphocyte antibody (CAMPATH-1). Lancet 2:483,1984 98. Welte K, Ciobanu N, Moore MA, et al: Defective interleukin-2 production in patients after bone marrow transplantation and in vitro restoration of defective T lymphocyte proliferation by highly purified interleukin-’2. Blood 64:380, 1984 99. Wexler LH, DeLaney TF, Tsokos M, et a1 Ifosfamide and etoposide plus vincristine, doxorubicm, and cyclophosphamide for newly diagnosed Ewing’s sarcoma family of tumors. Cancer 78:901, 1996 100. White CW, Wolf SJ, Korones DN, et al: Treatment of childhood angiomatous disease with recombinant interferon alfa-ZA. J Pediatr 118:59, 1991 101. Worth LL, Jaffe N, Benjamin R, et al: Phase I1 study of recombinant interleukin 1 a and etoposide in patients with relapsed osteosarcoma. Clin Cancer Res 3:1721, 1997 102. Worth LL, Jia S-F, An T, et al: ImmTher, a lipophilic derivative of muramly dipeptide, upregulates specific monocyte genes and activates monocyte-mediated tumoricidal activity Clin Immunol Immunother 48:312, 1999 103. Worth LL, Jia S-F, Zhou Z, et al: Intranasal therapy with an adenoviral vector containing the murine interleukin-12 gene eradicates osteosarcoma lung metastases. Clin Cancer Res 6:713,2000

Address reprint requests to Laura L. Worth, MD, PhD Department of Pediatrics, Box 87 The University of Texas The M. D. Anderson Cancer Center 1515 Holcombe Boulevard Houston, TX 77030